Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
AIDS ; 32(9): 1107-1114, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29596107

ABSTRACT

OBJECTIVE: In the post-HAART era, the incidence of some AIDS-defining cancers declined markedly likely reflecting HAART-related improvements in immunity, whereas incidence of some cancers such as cervical cancer has not been affected. Therefore, it is valuable to find whether antiretroviral drugs or prophylactic microbicides could treat or prevent these cancers, especially the cervical cancer. DESIGN: We screened the anti-HIV drugs, approved or in phase III clinical trials, to identify a potential anticancer drug candidate. METHODS: We chose cervical HeLa and SiHa cancer cells and focused on studying the antitumor effects in vitro and in vivo. Cell proliferation was measured by MTT assay, the cytotoxic effect was obtained through apoptosis as evidenced by Annexin V flow cytometry assay because of the arresting of cancer cells in G2/M phase of cell cycle. Nude mice xenograft model was performed to detect the antitumor effect in vivo. RESULTS: TMC120 was identified as a potential anticancer drug candidate. TMC120 displayed potent cytotoxic effect on various human cancer cells, including cervical carcinoma cell line HeLa and SiHa. Further mechanism study showed that TMC120 enhanced the polymerization of microtubules, which was followed by mitotic arrest, as well as abnormal mitotic spindles. TMC120 also substantially retarded the growth rate of the tumor in vivo. CONCLUSION: TMC120 is a potential chemoprophylactic and therapeutic agent for cervical cancers in a manner similar to paclitaxel, and could be suitable for helping healthy women to prevent HIV infection and cervical cancer.


Subject(s)
Anti-Retroviral Agents/pharmacology , Antineoplastic Agents/pharmacology , Microtubules/metabolism , Polymerization/drug effects , Pyrimidines/pharmacology , Animals , Anti-Retroviral Agents/administration & dosage , Antineoplastic Agents/administration & dosage , Apoptosis , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Evaluation, Preclinical , Female , Heterografts , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Neoplasms, Experimental/drug therapy , Pyrimidines/administration & dosage , Treatment Outcome
2.
Metab Brain Dis ; 33(1): 167-175, 2018 02.
Article in English | MEDLINE | ID: mdl-29101599

ABSTRACT

Anxiety disorder is a serious and burdensome psychiatric illness that frequently turn into chronic clinical conditions. Puerarin have been shown to be effective in the therapy of depression. However, few studies are concerned about the anxiolytic-like effects of puerarin. The current study aimed to evaluate the anxiolytic-like effects of puerarin and its possible mechanism. To evaluate this, the behavioral tests, i.e. Vogel-type conflict test (VTCT), elevated plus-maze test (EPMT), and open-field test (OFT) were conducted. Data showed that similar to the positive-control drug sertraline (Ser) (15 mg/kg, i.g.), the anxiolytic-like effects were produced by puerarin (60 and 120 mg/kg, i.g.) in VTCT and EMPT respectively without affecting locomotor activity in OFT. Moreover, the present study also found that consistent with Ser, the levels of allopregnanolone and serotonin (5-HT) in the prefrontal cortex and hippocampus were increased by puerarin (60 and 120 mg/kg, i.g.), respectively. In summary, the present study indicated that puerarin exerted the anxiolytic-like effects, which maybe associated with normalization of 5-HT levels and biosynthesis of allopregnanolone in brain.


Subject(s)
Anti-Anxiety Agents/pharmacology , Anxiety/drug therapy , Brain/metabolism , Isoflavones/pharmacology , Neurotransmitter Agents/metabolism , Animals , Brain/drug effects , Depression/drug therapy , Male , Maze Learning/drug effects , Pregnanolone/pharmacology , Rats, Sprague-Dawley , Serotonin/metabolism
3.
Sci Rep ; 7(1): 6266, 2017 07 24.
Article in English | MEDLINE | ID: mdl-28740098

ABSTRACT

The present study aimed to investigate the mechanisms underlying the antidepressant-like effects of puerarin via the chronic unpredictable stress (CUS) procedure in rats. Similar to Sertraline (Ser), Chronic treatment of puerarin (60 and 120 mg/kg, i.g) elicited the antidepressant-like effects by reversing the decreased sucrose preference in sucrose preference test (SPT), by blocking the increased latency to feed in novelty-suppressed feeding test (NSFT) and the increased immobility time in forced swimming test (FST) without affecting locomotor activity. However, acute puerarin treatment did not ameliorate the antidepressant- and anxiolytic- like effects in FST and NSFT, respectively. In addition, enzyme linked immunosorbent assay (ELISA) and high performance liquid chromatography-electrochemical detection (HPLC-ECD) showed that chronic treatment of puerarin (60 and 120 mg/kg, i.g) reversed the decreased levels of progesterone, allopregnanolone, serotonin (5-HT) and 5-Hydroxyindoleacetic acid (5-HIAA) in prefrontal cortex and hippocampus of post-CUS rats. Furthermore, puerarin (60 and 120 mg/kg, i.g) blocked the increased corticotropin releasing hormone (CRH), corticosterone (Cort) and adrenocorticotropic hormone (ACTH). Collectively, repeated administration of puerarin alleviated the behavioral deficits induced by chronic stress which was associated with the biosynthesis of neurosteroids, normalization of serotonergic system and preventing HPA axis dysfunction.


Subject(s)
Behavior, Animal/drug effects , Isoflavones/pharmacology , Stress, Psychological/complications , Stress, Psychological/drug therapy , Vasodilator Agents/pharmacology , Animals , Hypothalamo-Hypophyseal System/drug effects , Locomotion , Male , Pituitary-Adrenal System/drug effects , Rats , Rats, Sprague-Dawley , Stress, Psychological/physiopathology , Swimming
4.
Acta Pharmacol Sin ; 38(12): 1632-1641, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28713162

ABSTRACT

Both the anti- and pro-apoptotic members of the Bcl-2 family are regulated by a conserved Bcl-2 homology (BH3) domain. ABT-263 (Navitoclax), a novel BH3 mimetic and orally bioavailable Bcl-2 family inhibitor with high affinity for Bcl-xL, Bcl-2 and Bcl-w has entered clinical trials for cancer treatment. But the anticancer mechanisms of ABT-263 have not been fully elucidated. In this study we investigated the effects of ABT-263 on human esophageal cancer cells in vitro and to explore its anticancer mechanisms. Treatment with ABT-263 dose-dependently suppressed the viability of 3 human esophageal cancer cells with IC50 values of 10.7±1.4, 7.1±1.5 and 8.2±1.6 µmol/L, in EC109, HKESC-2 and CaES-17 cells, respectively. ABT-263 (5-20 µmol/L) dose-dependently induced G1/G0-phase arrest in the 3 cancer cell lines and induced apoptosis evidenced by increased the Annexin V-positive cell population and elevated levels of cleaved caspase 3, cleaved caspase 9 and PARP. We further demonstrated that ABT-263 treatment markedly increased the expression of p21Waf1/Cip1 and decreased the expression of cyclin D1 and phospho-Rb (retinoblastoma tumor suppressor protein) (Ser780) proteins that contributed to the G1/G0-phase arrest. Knockdown of p21Waf1/Cip1 attenuated ABT-263-induced G1/G0-phase arrest. Moreover, ABT-263 treatment enhanced pro-survival autophagy, shown as the increased LC3-II levels and decreased p62 levels, which counteracted its anticancer activity. Our results suggest that ABT-263 exerts cytostatic and cytotoxic effects on human esophageal cancer cells in vitro and enhances pro-survival autophagy, which counteracts its anticancer activity.


Subject(s)
Aniline Compounds/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Autophagy/drug effects , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/pathology , G1 Phase Cell Cycle Checkpoints/drug effects , Resting Phase, Cell Cycle/drug effects , Sulfonamides/pharmacology , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Structure-Activity Relationship , Tumor Cells, Cultured
5.
Oncotarget ; 7(12): 14693-707, 2016 Mar 22.
Article in English | MEDLINE | ID: mdl-26910910

ABSTRACT

Obatoclax, a pan-inhibitor of anti-apoptotic Bcl-2 proteins, exhibits cytotoxic effect on cancer cells through both apoptosis-dependent and -independent pathways. Here we show that obatoclax caused cytotoxicity in both cisplatin-sensitive and -resistant esophageal cancer cells. Although obatoclax showed differential apoptogenic effects in these cells, it consistently blocked autophagic flux, which was evidenced by concomitant accumulation of LC3-II and p62. Obatoclax was trapped in lysosomes and induced lysosome clustering. Obatoclax also substantially reduced the expression of lysosomal cathepsins B, D and L. Moreover, cathepsin knockdown was sufficient to induce cytotoxicity, connecting lysosomal function to cell viability. Consistent with the known function of autophagy, obatoclax caused the accumulation of polyubiquitinated proteins and showed synergy with proteasome inhibition. Taken together, our studies unveiled impaired lysosomal function as a novel mechanism whereby obatoclax mediates its cytotoxic effect in esophageal cancer cells.


Subject(s)
Apoptosis/drug effects , Cisplatin/pharmacology , Drug Resistance, Neoplasm/drug effects , Esophageal Neoplasms/pathology , Lysosomes/pathology , Pyrroles/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis Regulatory Proteins/metabolism , Autophagy/drug effects , Cathepsin B/metabolism , Cell Proliferation/drug effects , Enzyme Inhibitors/pharmacology , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/metabolism , Humans , Indoles , Lysosomes/drug effects , Lysosomes/metabolism , Tumor Cells, Cultured
6.
Eur J Pharm Sci ; 81: 137-48, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26474693

ABSTRACT

It has been demonstrated that COX-2-selective inhibitor celecoxib shows synergy with oxaliplatin for suppressing tumor growth. However, the benefit of adding celecoxib to oxaliplatin-based regimen in human esophageal cancer is largely unknown. In the present study, we demonstrated that celecoxib antagonized oxaliplatin-induced cytotoxicity and apoptosis independent of COX-2 inhibition in human esophageal cancer cells. Celecoxib decreased cellular oxaliplatin accumulation and Pt-DNA adduction formation due to reduced drug influx. Celecoxib alone or combined with oxaliplatin substantially reduced the expression of organic cation transporter 2 (OCT2). To this end, OCT2 knockdown was sufficient to reduce oxaliplatin uptake, connecting OCT2 expression to oxaliplatin accumulation. Moreover, oxaliplatin combined with celecoxib also showed no beneficial effect when compared with monotherapy in esophageal cancer cell-xenografted nude mice. To conclude, our data provide evidence that the addition of celecoxib to oxaliplatin-containing regimens for patients with OCT2-expressing cancers should be cautious.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Celecoxib/pharmacology , Cyclooxygenase 2 Inhibitors/pharmacology , Esophageal Neoplasms/metabolism , Organoplatinum Compounds/pharmacology , Animals , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Celecoxib/therapeutic use , Cell Line, Tumor , Cell Survival/drug effects , Cyclooxygenase 2 Inhibitors/therapeutic use , Drug Interactions , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/pathology , Female , Humans , Mice, Nude , Organic Cation Transport Proteins/genetics , Organic Cation Transport Proteins/metabolism , Organic Cation Transporter 2 , Organoplatinum Compounds/therapeutic use , Oxaliplatin , RNA, Small Interfering/genetics , Tumor Burden/drug effects
7.
Cancer Lett ; 355(1): 34-45, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25236911

ABSTRACT

Cisplatin-based chemotherapy frequently resulted in acquired resistance. The underpinning mechanism of such resistance remains obscure especially in relation to autophagic response. This study thus investigated the role of autophagy in the anticancer activity of cisplatin in human esophageal cancer cells with acquired cisplatin resistance. In response to cisplatin treatment, EC109 cells exhibited substantial apoptosis and senescence whereas cisplatin-resistant EC109/CDDP cells exhibited resistance. In this respect, cisplatin increased ERK phosphorylation whose inhibition by MEK inhibitor significantly attenuated the cytotoxic and cytostatic effect of cisplatin. Notably, cisplatin preferentially induces autophagy in EC109/CDDP cells but not in EC109 cells. Moreover, the induction of autophagy was accompanied by the suppression of mTORC1 activity. Abolition of autophagy by pharmacological inhibitors or knockdown of ATG5/7 re-sensitized EC109/CDDP cells. Co-administration of an autophagy inhibitor chloroquine and cisplatin significantly suppressed tumor growth whereas cisplatin monotherapy failed to elicit anticancer activity in nude mice xenografted with EC109/CDDP cells. To conclude, our data implicate autophagic response as a key mechanism of acquired resistance to cisplatin, suggesting that autophagy is a novel target to improve therapy efficiency of cisplatin toward human esophageal cancers with acquired resistance.


Subject(s)
Antineoplastic Agents/pharmacology , Autophagy/drug effects , Chloroquine/pharmacology , Cisplatin/pharmacology , Drug Resistance, Neoplasm , Esophageal Neoplasms/drug therapy , Animals , Autophagy-Related Protein 5 , Autophagy-Related Protein 7 , Cell Line, Tumor , Cellular Senescence/drug effects , Dose-Response Relationship, Drug , Enzyme Activation , Esophageal Neoplasms/genetics , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Humans , Mechanistic Target of Rapamycin Complex 1 , Mice, Inbred BALB C , Mice, Nude , Microtubule-Associated Proteins/antagonists & inhibitors , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Multiprotein Complexes/metabolism , Phosphorylation , Protein Kinase Inhibitors/pharmacology , RNA Interference , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Time Factors , Transfection , Tumor Burden/drug effects , Ubiquitin-Activating Enzymes/antagonists & inhibitors , Ubiquitin-Activating Enzymes/genetics , Ubiquitin-Activating Enzymes/metabolism , Xenograft Model Antitumor Assays
8.
Nan Fang Yi Ke Da Xue Xue Bao ; 34(6): 792-7, 2014 Jun.
Article in Chinese | MEDLINE | ID: mdl-24968832

ABSTRACT

OBJECTIVE: To explore the antagonizing effect of celecoxib against the cytotoxicity of carboplatin in human esophageal cancer cells. METHODS: The cell viability of cisplatin-resistant cell line EC109/CDDP and its parental cell line EC109 exposed to carboplatin alone or carboplatin plus celecoxib was determined by MTT assay. The expression of CTR1, caspase-3 activation and PARP cleavage in the exposed cells were examined by Western blotting. Caspase-3 activity and cell apoptosis after the exposure were detected with Caspase-3/7 assay and flow cytometry, respectively. The effect of celecoxib on carboplatin accumulation in the cells was measured using inductively coupled plasma mass spectrometry (ICP-MS). RESULTS: Celecoxib treatment significantly increased the IC50 of carboplatin, suppressed carboplatin-induced caspase-3 and PARP cleavage and caspase-3 activity in EC109 and EC109/CDDP cells. Celecoxib also inhibited carboplatin-induced apoptosis and suppressed intracellular carboplatin accumulation in both cell lines. A combined exposure to celecoxib and carboplatin did not cause significant changes in the protein expression of CTR1. CONCLUSION: Celecoxib antagonizes the cytotoxic effect of carboplatin and inhibits carboplatin-induced apoptosis in human esophageal cancer cells by reducing intracellular carboplatin accumulation.


Subject(s)
Carboplatin/antagonists & inhibitors , Esophageal Neoplasms/pathology , Pyrazoles/pharmacology , Sulfonamides/pharmacology , Apoptosis , Blotting, Western , Caspase 3/metabolism , Celecoxib , Cell Line, Tumor/drug effects , Cell Survival , Drug Interactions , Esophageal Neoplasms/metabolism , Humans
9.
J Cell Biochem ; 115(9): 1624-35, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24788582

ABSTRACT

Pan-Bcl-2 family inhibitor obatoclax has been demonstrated to be effective against various cancers, of which the mechanism of action is not fully understood. In this study, we demonstrate that obatoclax suppressed esophageal cancer cell viability with concomitant G1/G0-phase cell cycle arrest. At the tested concentrations (1/2 IC50 and IC50), obatoclax neither induced PARP cleavage nor increased the Annexin V-positive population, suggesting G1/G0-phase arrest rather than apoptosis accounts for most of the reduction of cell viability produced by obatoclax. Double knockdown of Bak and Bax by small interference RNA failed to block obatoclax-induced G1/G0-phase arrest, implying its role in cell cycle progression is Bak/Bax-independent. The cell cycle arresting effect of obatoclax was associated with up-regulation of p21(waf1/Cip1). Knockdown of p21(waf1/Cip1) significantly attenuated obatoclax-induced G1/G0-phase arrest. Although obatoclax stimulated phosphorylation of Erk, p38, and JNK, pharmacological inhibition of p38 but not Erk or JNK blocked obatoclax-induced G1/G0-phase arrest. Moreover, knockdown of p38 abolished the cell cycle arresting effect of obatoclax. In consistent with this finding, inhibition of p38 blocked obatoclax-induced p21(waf1/Cip1) expression while inhibition of Erk or JNK failed to exert similar effect. To conclude, these findings suggest that obatoclax induced cell cycle arrest via p38/p21(waf1/Cip1) signaling pathway. This study may shed a new light on the anti-cancer activity of obatoclax in relation to cell cycle arrest.


Subject(s)
Antineoplastic Agents/pharmacology , MAP Kinase Signaling System/drug effects , Pyrroles/pharmacology , bcl-2 Homologous Antagonist-Killer Protein/antagonists & inhibitors , bcl-2-Associated X Protein/antagonists & inhibitors , Cell Cycle/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Humans , Indoles , Phosphorylation , p38 Mitogen-Activated Protein Kinases/metabolism
10.
Eur J Pharmacol ; 729: 22-9, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24462571

ABSTRACT

The wide range of inflammation mechanisms under control by NF-κB makes this pathway as an attractive target for new anti-inflammatory drugs. Herein, we showed that a new dibenzocyclooctadiene lignan analog XLYF-104-6, with a chemical name of 1,2,3,10,11-pentamethoxydibenzocycloocta-6,7-[c] pyrrole-1,3-dione, inhibited lipopolysaccharide (LPS)-induced NF-κB activation in RAW264.7 cells through preventing IκBα degradation and p65 nuclear translocation. The inhibitory activity of this compound on NF-κB activation contributes to the reduction of LPS-induced TNF-α and IL-6 productions. Notably, XLYF-104-6 suppressed LPS-induced iNOS expression and NO production in a NF-κB independent manner, since IKK inhibitor BAY 11-7082 has failed to exert similar inhibitory effect on iNOS expression and NO production. In addition, XLFY-104-6 also exerted anti-inflammatory action in endotoxemic mice by decreasing plasma LPS-induced TNF-α and IL-1ß levels as well as increasing plasma LPS-induced IL-10 concentrations. These findings suggest XLYF-104-6 could act as a leading compound for developing a potential anti-inflammatory drug.


Subject(s)
Cyclooctanes/therapeutic use , Inflammation Mediators/metabolism , Lignans/therapeutic use , Lipopolysaccharides/toxicity , Macrophages/metabolism , Sepsis/metabolism , Sepsis/prevention & control , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Cell Line , Cyclooctanes/chemistry , Cyclooctanes/pharmacology , Dose-Response Relationship, Drug , Female , Inflammation Mediators/antagonists & inhibitors , Lignans/chemistry , Lignans/pharmacology , Macrophages/drug effects , Male , Mice , Mice, Inbred BALB C , Sepsis/chemically induced
11.
Cancer Lett ; 329(2): 189-96, 2013 Feb 28.
Article in English | MEDLINE | ID: mdl-23142285

ABSTRACT

Cisplatin is a chemotherapeutic drug widely used for the treatment of gastric cancer. The benefit of including COX-2-selective inhibitors in cisplatin-based regimens on anti-cancer effect remains uncertain. In the present study, celecoxib and SC-236 antagonized cisplatin-induced cytotoxicity and apoptosis, whereas indomethancin and nimesulide exerted no effect, implying a COX-2-independent mechanism. Celecoxib decreased whole-cell cisplatin accumulation and DNA platination, resulting from reduced influx. In addition, combined treatment did not elicit greater antitumor activity than cisplatin or celecoxib monotherapy in vivo in a gastric xenograft model. Therefore, treatment strategies with celecoxib in combination with cisplatin should act cautiously.


Subject(s)
Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , Cyclooxygenase 2 Inhibitors/pharmacology , Pyrazoles/pharmacology , Sulfonamides/pharmacology , Animals , Antineoplastic Agents/metabolism , Apoptosis/drug effects , Biological Transport/drug effects , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Celecoxib , Cell Line, Tumor , Cell Survival/drug effects , Cisplatin/metabolism , Copper Transporter 1 , Cyclooxygenase 2/metabolism , DNA Adducts/metabolism , Dinoprostone/metabolism , Drug Interactions , Female , Gene Expression/drug effects , Humans , Inhibitory Concentration 50 , Intracellular Fluid/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...