Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Bioorg Chem ; 107: 104395, 2021 02.
Article in English | MEDLINE | ID: mdl-33384144

ABSTRACT

We report the design, synthesis, and biological evaluation of 13 new and 1 known anthraquinone derivatives which exerted cytotoxicity against PC3, A549 and NTUB1 cell lines. The results indicate that, among these 14, compounds-1 and 14 showed the highest growth inhibitory effect on NTUB1 and PC3 cells, respectively. Compound-1 at lower doses targets DNA, induces DNA damage and subsequently triggers G2/M arrest and apoptotic cell death at 24 h. Previously we reported that 14 induced PC3 cell autophagy and in treated PC3 cells, cleaved caspase-3 and cleaved PARP, and survivin did not increase and increase, respectively. The autophagic and necrotic cell deaths mediated by 14-triggered ROS generation. Our study is the first to investigate the biological mechanism of 14 action in detail. We find that when 14 was co-administrated with Bafilomycin A1 (BAF) in PC3 cells, rapid necrotic cell death occurred with no cleaved caspase-3 and cleaved PARP activation and increasing the expression of survivin. We further show that necrotic signaling in these cells coincided with production of reactive oxygen species. In the present study, we developed methods to synthesize five new 14 analogues for studing the structure-activity relationships. This study could provide valuable sight to find new antitumor agents for cancer therapy.


Subject(s)
Anthraquinones/pharmacology , Antineoplastic Agents/pharmacology , Anthraquinones/chemical synthesis , Antineoplastic Agents/chemical synthesis , Apoptosis/drug effects , Autophagy/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , DNA Damage/drug effects , Drug Design , Drug Screening Assays, Antitumor , G2 Phase Cell Cycle Checkpoints/drug effects , Humans , Molecular Structure , Reactive Oxygen Species/metabolism , Structure-Activity Relationship
2.
J Nat Prod ; 80(5): 1421-1427, 2017 05 26.
Article in English | MEDLINE | ID: mdl-28445049

ABSTRACT

The tree Aquilaria malaccensis is a valuable source of agarwood, which is used in herbal medicinal preparations. Phytochemical research on A. malaccensis seeds has led to the isolation of four new phorbol esters (1-4), two known phorbol esters (5, isolated from Nature for the first time, and 6), and two known glycerides (7 and 8). The structures of these isolates were elucidated by means of spectroscopic data interpretation. The inflammation-modulatory activities of the isolates on elastase release and superoxide anion generation in human neutrophils were evaluated. Interestingly, phorbol esters 1, 5, and 6 showed potent inhibitory activity on elastase release in human neutrophils, with IC50 values of 2.7, 0.8, and 2.1 µM, respectively. All isolated phorbol esters exerted enhancing activity on superoxide anion generation. The results indicated that phorbol esters may play a bilateral modulatory role in the processes of inflammation. In addition, the compounds were evaluated for their cytotoxic properties against HepG2 (hepatoma), MDA-MB-231 (breast), and A549 (lung) cancer cells, but all compounds were inactive for all cell lines used (IC50 > 10 µM).


Subject(s)
Anti-Inflammatory Agents/isolation & purification , Anti-Inflammatory Agents/pharmacology , Glycerides/isolation & purification , Glycerides/pharmacology , Neutrophils/drug effects , Phorbol Esters/isolation & purification , Phorbol Esters/pharmacology , Seeds/chemistry , Thymelaeaceae/chemistry , Anti-Inflammatory Agents/chemistry , Glycerides/chemistry , Humans , Molecular Structure , Neutrophils/chemistry , Phorbol Esters/chemistry
3.
Semin Cancer Biol ; 35 Suppl: S78-S103, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25936818

ABSTRACT

Apoptosis or programmed cell death is natural way of removing aged cells from the body. Most of the anti-cancer therapies trigger apoptosis induction and related cell death networks to eliminate malignant cells. However, in cancer, de-regulated apoptotic signaling, particularly the activation of an anti-apoptotic systems, allows cancer cells to escape this program leading to uncontrolled proliferation resulting in tumor survival, therapeutic resistance and recurrence of cancer. This resistance is a complicated phenomenon that emanates from the interactions of various molecules and signaling pathways. In this comprehensive review we discuss the various factors contributing to apoptosis resistance in cancers. The key resistance targets that are discussed include (1) Bcl-2 and Mcl-1 proteins; (2) autophagy processes; (3) necrosis and necroptosis; (4) heat shock protein signaling; (5) the proteasome pathway; (6) epigenetic mechanisms; and (7) aberrant nuclear export signaling. The shortcomings of current therapeutic modalities are highlighted and a broad spectrum strategy using approaches including (a) gossypol; (b) epigallocatechin-3-gallate; (c) UMI-77 (d) triptolide and (e) selinexor that can be used to overcome cell death resistance is presented. This review provides a roadmap for the design of successful anti-cancer strategies that overcome resistance to apoptosis for better therapeutic outcome in patients with cancer.


Subject(s)
Apoptosis/genetics , Molecular Targeted Therapy , Neoplasms/drug therapy , Neoplasms/genetics , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Autophagy/genetics , Cell Proliferation/genetics , Drug Resistance, Neoplasm/genetics , Humans , Neoplasms/pathology , Signal Transduction/drug effects , Signal Transduction/genetics
4.
Int J Mol Sci ; 17(3): 398, 2016 Mar 21.
Article in English | MEDLINE | ID: mdl-27007372

ABSTRACT

The Aquilaria malaccensis (Thymelaeaceae) tree is a source of precious fragrant resin, called agarwood, which is widely used in traditional medicines in East Asia against diseases such as asthma. In our continuous search for active natural products, A. malaccensis seeds ethanolic extract demonstrated antiallergic effect with an IC50 value less than 1 µg/mL. Therefore, the present research aimed to purify and identify the antiallergic principle of A. malaccensis through a bioactivity-guided fractionation approach. We found that phorbol ester-rich fraction was responsible for the antiallergic activity of A. malaccensis seeds. One new active phorbol ester, 12-O-(2Z,4E,6E)-tetradeca-2,4,6-trienoylphorbol-13-acetate, aquimavitalin (1) was isolated. The structure of 1 was assigned by means of 1D and 2D NMR data and high-resolution mass spectrometry (HR-MS). Aquimavitalin (1) showed strong inhibitory activity in A23187- and antigen-induced degranulation assay with IC50 values of 1.7 and 11 nM, respectively, with a therapeutic index up to 71,000. The antiallergic activities of A. malaccensis seeds and aquimavitalin (1) have never been revealed before. The results indicated that A. malaccensis seeds and the pure compound have the potential for use in the treatment of allergy.


Subject(s)
Anti-Allergic Agents/chemistry , Phorbol Esters/chemistry , Plant Extracts/chemistry , Thymelaeaceae/chemistry , Animals , Anti-Allergic Agents/pharmacology , Cell Line, Tumor , Phorbol Esters/pharmacology , Plant Extracts/pharmacology , Rats , Seeds/chemistry
5.
Carcinogenesis ; 36 Suppl 1: S184-202, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26106137

ABSTRACT

One of the important 'hallmarks' of cancer is angiogenesis, which is the process of formation of new blood vessels that are necessary for tumor expansion, invasion and metastasis. Under normal physiological conditions, angiogenesis is well balanced and controlled by endogenous proangiogenic factors and antiangiogenic factors. However, factors produced by cancer cells, cancer stem cells and other cell types in the tumor stroma can disrupt the balance so that the tumor microenvironment favors tumor angiogenesis. These factors include vascular endothelial growth factor, endothelial tissue factor and other membrane bound receptors that mediate multiple intracellular signaling pathways that contribute to tumor angiogenesis. Though environmental exposures to certain chemicals have been found to initiate and promote tumor development, the role of these exposures (particularly to low doses of multiple substances), is largely unknown in relation to tumor angiogenesis. This review summarizes the evidence of the role of environmental chemical bioactivity and exposure in tumor angiogenesis and carcinogenesis. We identify a number of ubiquitous (prototypical) chemicals with disruptive potential that may warrant further investigation given their selectivity for high-throughput screening assay targets associated with proangiogenic pathways. We also consider the cross-hallmark relationships of a number of important angiogenic pathway targets with other cancer hallmarks and we make recommendations for future research. Understanding of the role of low-dose exposure of chemicals with disruptive potential could help us refine our approach to cancer risk assessment, and may ultimately aid in preventing cancer by reducing or eliminating exposures to synergistic mixtures of chemicals with carcinogenic potential.


Subject(s)
Carcinogenesis/chemically induced , Carcinogens, Environmental/adverse effects , Environmental Exposure/adverse effects , Hazardous Substances/adverse effects , Neoplasms/chemically induced , Neoplasms/etiology , Neovascularization, Pathologic/chemically induced , Animals , Humans
6.
Toxicol Appl Pharmacol ; 264(1): 104-13, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-22842013

ABSTRACT

Quercetin is a bioflavonoid that exhibits several biological functions in vitro and in vivo. Quercetin 3-O-methyl ether (Q3) is a natural product reported to have pharmaceutical activities, including antioxidative and anticancer activities. However, little is known about the mechanism by which it protects cells from oxidative stress. This study was designed to investigate the mechanisms by which Q3 protects against Cu(2+)-induced cytotoxicity. Exposure to Cu(2+) resulted in the death of mouse liver FL83B cells, characterized by apparent apoptotic features, including DNA fragmentation and increased nuclear condensation. Q3 markedly suppressed Cu(2+)-induced apoptosis and mitochondrial dysfunction, characterized by reduced mitochondrial membrane potential, caspase-3 activation, and PARP cleavage, in Cu(2+)-exposed cells. The involvement of PI3K, Akt, Erk, FOXO3A, and Mn-superoxide dismutase (MnSOD) was shown to be critical to the survival of Q3-treated FL83B cells. The liver of both larval and adult zebrafish showed severe damage after exposure to Cu(2+) at a concentration of 5µM. Hepatic damage induced by Cu(2+) was reduced by cotreatment with Q3. Survival of Cu(2+)-exposed larval zebrafish was significantly increased by cotreatment with 15µM Q3. Our results indicated that Cu(2+)-induced apoptosis in FL83B cells occurred via the generation of ROS, upregulation and phosphorylation of Erk, overexpression of 14-3-3, inactivation of Akt, and the downregulation of FOXO3A and MnSOD. Hence, these results also demonstrated that Q3 plays a protective role against oxidative damage in zebrafish liver and remarked the potential of Q3 to be used as an antioxidant for hepatocytes.


Subject(s)
Antioxidants/pharmacology , Copper/toxicity , Liver/drug effects , Oxidative Stress/drug effects , Quercetin/analogs & derivatives , Animals , Apoptosis/drug effects , Cell Line , DNA Fragmentation/drug effects , Down-Regulation/drug effects , Liver/cytology , Liver/pathology , MAP Kinase Signaling System/drug effects , Membrane Potential, Mitochondrial/drug effects , Mice , Mitochondria, Liver/drug effects , Mitochondria, Liver/pathology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Quercetin/pharmacology , Reactive Oxygen Species/metabolism , Up-Regulation/drug effects , Zebrafish
7.
Article in English | MEDLINE | ID: mdl-23091557

ABSTRACT

Plants are an invaluable source of potential new anti-cancer drugs. Momordica charantia is one of these plants with both edible and medical value and reported to exhibit anticancer activity. To explore the potential effectiveness of Momordica charantia, methanol extract of Momordica charantia (MCME) was used to evaluate the cytotoxic activity on four human cancer cell lines, Hone-1 nasopharyngeal carcinoma cells, AGS gastric adenocarcinoma cells, HCT-116 colorectal carcinoma cells, and CL1-0 lung adenocarcinoma cells, in this study. MCME showed cytotoxic activity towards all cancer cells tested, with the approximate IC(50) ranging from 0.25 to 0.35 mg/mL at 24 h. MCME induced cell death was found to be time-dependent in these cells. Apoptosis was demonstrated by DAPI staining and DNA fragmentation analysis using agarose gel electrophoresis. MCME activated caspase-3 and enhanced the cleavage of downstream DFF45 and PARP, subsequently leading to DNA fragmentation and nuclear condensation. The apoptogenic protein, Bax, was increased, whereas Bcl-2 was decreased after treating for 24 h in all cancer cells, indicating the involvement of mitochondrial pathway in MCME-induced cell death. These findings indicate that MCME has cytotoxic effects on human cancer cells and exhibits promising anti-cancer activity by triggering apoptosis through the regulation of caspases and mitochondria.

8.
Article in English | MEDLINE | ID: mdl-23320038

ABSTRACT

Momordica charantia has been found to exhibit anticancer activity, in addition to its well-known therapeutic functions. We have demonstrated that the leaf extract of Momordica charantia (MCME) induces apoptosis in several human cancer cells through caspase- and mitochondria-dependent pathways. In this study, a different susceptibility to MCME was found in human lung adenocarcinoma CL1 cells with different metastatic ability, leading to the significant difference of cell viability and invasiveness between MCME-treated CL1-0 and CL1-5 cells. MCME was found to upregulate the expression of Wnt-2 and affect the migratory and invasive ability of CL1 cells through suppressed MMP-2 and MMP-9 enzymatic activities. We proposed that MCME mediates inhibition against migration of CL1 cells by reducing the expression and activation of Src and FAK to decrease the expression of downstream Akt, ß-catenin, and MMPs.

9.
Molecules ; 17(8): 9348-60, 2012 Aug 03.
Article in English | MEDLINE | ID: mdl-22864242

ABSTRACT

Corchorus olitorius L.,is a culinary and medicinal herb, widely used as a vegetable in several countries in Asia. Many studies have shown that C. olitorius contains several antioxidants and exhibits anti-inflammatory and anti-proliferative activities in various in vitro and in vivo settings. Recently, C. olitorius has been approved for its antitumor activity; however, the underlying molecular mechanisms remain unclear. The goal of this study was to investigate the effects of ethanol extract of C. olitorius (ECO) on the growth of human hepatocellular carcinoma (HepG2) cells and gain some insights into the underlying mechanisms of its action. We found that HepG2 cells, treated with ECO for 24 h at a concentration higher than 12.5 µg/mL, displayed a strong reduction in cell viability, whereas normal FL83B hepatocytes were not affected. DNA fragmentation and nuclear condensation were evidenced by the increased subG1 population of ECO-treated HepG2 cells. ECO triggered the activation of procaspases-3 and -9 and caused the cleavage of downstream substrate, poly ADP-ribose polymerase (PARP), followed by down-regulation of the inhibitor of caspase-activated DNase (ICAD) signaling. Moreover, the increased release of cytochrome c from mitochondria with decreased membrane potential demonstrated the apoptosis induced through the caspases cascade. Our findings indicated that ECO might be effective against hepatocellular carcinoma through induction of apoptosis via mitochondria-dependent pathway.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Corchorus/chemistry , Mitochondria/drug effects , Plant Extracts/pharmacology , Plant Leaves/chemistry , Antineoplastic Agents/isolation & purification , Apoptosis Regulatory Proteins/metabolism , Carcinoma, Hepatocellular/drug therapy , Cell Cycle/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , DNA Fragmentation , Drug Screening Assays, Antitumor , Ethanol/chemistry , Hep G2 Cells , Hepatocytes/drug effects , Humans , Liver Neoplasms/drug therapy , Plant Extracts/isolation & purification , Solvents/chemistry
10.
Molecules ; 16(4): 3212-21, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21499221

ABSTRACT

The ability of the flavan kazinol Q (KQ) to induce DNA breakage in the presence of Cu(II) was examined by agarose gel electrophoresis using supercoiled plasmid DNA. In KQ-mediated DNA breakage reaction, the involvement of reactive oxygen species (ROS), H(2)O(2) and O(2)- was established by the inhibition of DNA breakage by catalase and revealed DNA breakage by superoxide dismutase (SOD). The cell viability of gastric carcinoma SCM-1 cells treated with various concentrations of KQ was significantly decreased by cotreatment with Cu(II). Treatment of SCM-1 cells with 300 µM Cu(II) enhanced the necrosis induced by 100 µM KQ. Treatment of SCM-1 cells with 100 mM KQ in the presence of 300 mM Cu(II) increased the generation of H(2)O(2). Taken together, the above finding suggested that KQ cotreatment with Cu(II) produced increased amounts of H(2)O(2), thus enhancing subsequent cell death due to necrosis.


Subject(s)
Broussonetia/chemistry , Cell Death/drug effects , Copper/pharmacology , Flavonoids/pharmacology , Hemiterpenes/pharmacology , Reactive Oxygen Species/metabolism , Cell Line, Tumor , DNA Damage , Humans , Plasmids
11.
Cancers (Basel) ; 13(1)2021 Jan 04.
Article in English | MEDLINE | ID: mdl-33406633

ABSTRACT

Oncolytic viruses (OVs) and phytochemical ursolic acid (UA) are two efficacious therapeutic candidates in development against breast cancer, the deadliest women's cancer worldwide. However, as single agents, OVs and UA have limited clinical efficacies. As a common strategy of enhancing monotherapeutic anticancer efficacy, we explored the combinatorial chemovirotherapeutic approach of combining oncolytic measles virus (MV), which targets the breast tumor marker Nectin-4, and the anticancer UA against breast adenocarcinoma. Our findings revealed that in vitro co-treatment with UA synergistically potentiated the killing of human breast cancer cells by oncolytic MV, without UA interfering the various steps of the viral infection. Mechanistic studies revealed that the synergistic outcome from the combined treatment was mediated through UA's potentiation of apoptotic killing by MV. To circumvent UA's poor solubility and bioavailability and strengthen its clinical applicability, we further developed UA nanoparticles (UA-NP) by nanoemulsification. Compared to the non-formulated UA, UA-NP exhibited improved drug dissolution property and similarly synergized with oncolytic MV in inducing apoptotic breast cancer cell death. This oncolytic potentiation was partly attributed to the enhanced autophagic flux induced by the UA-NP and MV combined treatment. Finally, the synergistic effect from the UA-NP and MV combination was also observed in BT-474 and MDA-MB-468 breast cancer cells. Our study thus highlights the potential value of oncolytic MV and UA-based chemovirotherapy for further development as a treatment strategy against breast cancer, and the feasibility of employing nanoformulation to enhance UA's applicability.

12.
Cells ; 8(4)2019 03 29.
Article in English | MEDLINE | ID: mdl-30934919

ABSTRACT

Mitophagy is a selective form of autophagy, targeting damaged mitochondria for lysosomal degradation. Although HCV infection has been shown to induce mitophagy, the precise underlying mechanism and the effector protein responsible remain unclear. Herein, we demonstrated that the HCV non-structural protein 5A (NS5A) plays a key role in regulating cellular mitophagy. Specifically, the expression of HCV NS5A in the hepatoma cells triggered hallmarks of mitophagy including mitochondrial fragmentation, loss of mitochondrial membrane potential, and Parkin translocation to the mitochondria. Furthermore, mitophagy induction through the expression of NS5A led to an increase in autophagic flux as demonstrated by an accumulation of LC3II in the presence of bafilomycin and a time-dependent decrease in p62 protein level. Intriguingly, the expression of NS5A concomitantly enhanced reactive oxygen species (ROS) production, and treatment with an antioxidant attenuated the NS5A-induced mitophagy event. These phenomena are similarly recapitulated in the NS5A-expressing HCV subgenomic replicon cells. Finally, we demonstrated that expression of HCV core, which has been documented to inhibit mitophagy, blocked the mitophagy induction both in cells harboring HCV replicating subgenomes or expressing NS5A alone. Our results, therefore, identified a new role for NS5A as an important regulator of HCV-induced mitophagy and have implications to broadening our understanding of the HCV-mitophagy interplay.


Subject(s)
Hepacivirus/metabolism , Mitochondrial Dynamics , Mitophagy , Viral Nonstructural Proteins/metabolism , Autophagy , Cell Line, Tumor , Humans , Lipids/chemistry , Membrane Potential, Mitochondrial , Microtubule-Associated Proteins/metabolism , Mitochondria/metabolism , Protein Transport , Reactive Oxygen Species/metabolism , Replicon/genetics , Ubiquitin-Protein Ligases/metabolism
13.
Mol Cells ; 26(4): 396-403, 2008 Oct 31.
Article in English | MEDLINE | ID: mdl-18612239

ABSTRACT

The first ORF of the ARV S1133 S1 segment encodes the nonstructural protein p10, which is responsible for the induction of cell syncytium formation. However, p10-dependent signaling during syncytium formation is fully unknown. Here, we show that dominant negative RhoA, Rho inhibitor C3 exoenzyme, ROCK/Rho-kinase inhibitor Y-27632 and Rac1 inhibitor NSC23766 inhibit p10-mediated cell fusion. p10 over-expression is concomitant with activation and membrane translocation of RhoA and Rac1, but not cdc42. RhoA and Rac1 downstream events, including JNK phosphorylation and transcription factor AP-1 and NF-kappaB activation, as well as MLC expression and phosphorylation are simultaneously activated by p10. p10 point mutant T13M possessed 20% fusion-inducing ability and four p10 fusion-deficient mutants V15M, V19M, C21S and L32A reduced or lost their ability to activate RhoA and Rac1 signaling. We conclude that p10-mediated syncytium formation proceeds by utilizing RhoA and Rac1-dependent signaling.


Subject(s)
Giant Cells/metabolism , Orthoreovirus, Avian/metabolism , Viral Proteins/metabolism , rac1 GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/metabolism , Amino Acid Sequence , Animals , Chlorocebus aethiops , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Giant Cells/drug effects , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Molecular Sequence Data , Mutant Proteins/chemistry , Mutant Proteins/metabolism , NF-kappa B/metabolism , Point Mutation/drug effects , Signal Transduction/drug effects , Transcription Factor AP-1/metabolism , Vero Cells , Viral Proteins/chemistry , rac1 GTP-Binding Protein/antagonists & inhibitors , rhoA GTP-Binding Protein/antagonists & inhibitors
14.
Food Chem Toxicol ; 46(7): 2394-401, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18462856

ABSTRACT

The ability of flavonoid glycosides isolated from several plants to induce DNA breakage was examined using supercoiled plasmid pBR322 DNA by agarose gel electrophoresis in the presence of Cu(II). Among all the compounds, 1, 4, and 6 could cause significant breakages of supercoiled plasmid pBR322 DNA in the presence of Cu(II). Cu(I) was not shown to be an essential intermediate in the process of pBR322 DNA breakage by using the Cu(I)-specific sequestering reagent neocuproine. A decreased cell viability was enhanced in gastric carcinoma SCM-1 cells treating with lower concentrations of 1 and 6 when cotreated with increased concentrations of Cu(II), respectively. Treatments of SCM-1 cells with 500 microM of 1 in the presence of 300 or 500 microM of Cu(II) inhibited the Cu(II)-induced apoptosis. Compound 1 (500 microM) could prevent cell death by inhibiting the 500 microM Cu(II)-induced apoptosis and necrosis, but did not have any effect on the mitochondrial membrane potential changed by 500 microM Cu(II). Both compounds 1 and 6 could inhibit the DNA breakages caused by O2- while 1 also revealed inhibitory effect on xanthine oxidase with an IC50 value of 22.7+/-6.9 microM. These results indicated that compound 1 with a higher concentration may probably mediate through the suppression of xanthine oxidase activity and reduce reactive oxygen species (ROS) induced by high concentration of Cu(II) (500 microM) and prevent the following cell death.


Subject(s)
Apoptosis/drug effects , Copper/pharmacology , DNA Damage/drug effects , DNA, Superhelical/drug effects , Flavonoids/pharmacology , Glycosides/pharmacology , Cell Survival/drug effects , Copper/chemistry , DNA Breaks/drug effects , Dose-Response Relationship, Drug , Drug Interactions , Electrophoresis, Agar Gel , Flavonoids/chemistry , Free Radical Scavengers , Free Radicals , Glycosides/chemistry , Humans , Membrane Potentials/drug effects , Oxidation-Reduction , Plasmids , Tumor Cells, Cultured , Xanthine Oxidase/antagonists & inhibitors
15.
Chem Biol Interact ; 281: 60-68, 2018 Feb 01.
Article in English | MEDLINE | ID: mdl-29273567

ABSTRACT

The autophagy of human prostate cancer cells (PC3 cells) induced by a new anthraquinone derivative, 1-Hydroxy-3-[(E)-4-(piperazine-diium)but-2-enyloxy]-9,10-anthraquinone ditrifluoroactate (PA) was investigated, and the relationship between autophagy and reactive oxygen species (ROS) generation was studied. The results indicated that PA induced PC3 cell death in a time- and dose-dependent manner, could inhibit PC3 cell growth by G1 phase cell cycle arrest and corresponding decrease in the G2/M cell population and induced S-phase arrest accompanied by a significant decrease G2/M and G1 phase numbers after PC3 cells treated with PA for 48 h, and increased the accumulation of autophagolysosomes and microtubule-associated protein LC3-ll, a marker of autophagy. However, these phenomenon were not observed in the group pretreated with the autophagy inhibitor 3-MA or Bafilomycin A1 (BAF), suggesting that PA induced PC3 cell autophagy. In addition, we found that PA triggered ROS generation in cells, while the levels of ROS decreased in the N-acetylcysteine (NAC) co-treatment, indicating that PA-mediated autophagy was partly blocked by NAC. In summary, the autophagic cell death of human PC3 cells mediated by PA-triggered ROS generation.


Subject(s)
Anthraquinones/toxicity , Apoptosis/drug effects , Autophagy/drug effects , Acetylcysteine/pharmacology , Adenine/analogs & derivatives , Adenine/pharmacology , Anthraquinones/chemical synthesis , Anthraquinones/chemistry , Caspase 3/metabolism , Cell Line, Tumor , G1 Phase Cell Cycle Checkpoints/drug effects , Humans , Macrolides/pharmacology , Microscopy, Electron, Transmission , Microtubule-Associated Proteins/metabolism , Reactive Oxygen Species/metabolism
16.
Cancer Lett ; 257(2): 252-62, 2007 Nov 18.
Article in English | MEDLINE | ID: mdl-17881119

ABSTRACT

Prostate apoptosis response-4 (Par-4) is a proapoptotic gene that selectively induces cell death in most cancer cells. In addition to the increased percentage of apoptotic cells, caspase-3 activity, and poly (ADP-ribose) polymerase (PARP) cleavage, we demonstrate that elevated expression of Par-4 and nuclear entry resulted in apoptosis of nasopharyngeal carcinoma (NPC) cell lines either in serum deprivation or by ectopic overexpression of Par-4. Moreover, disassociation from the Par-4/Akt complex was correlated with the induced proapoptotic ability of Par-4. Therefore, our data suggest that the cytoplasmic localization and expression level of endogenous Par-4 in NPC cells are not sufficient to augment apoptosis.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis/physiology , Intracellular Fluid/metabolism , Apoptosis/drug effects , Apoptosis/genetics , Apoptosis Regulatory Proteins/genetics , Blotting, Western , Caspase 3/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Survival/drug effects , Cell Survival/genetics , Cell Survival/physiology , Culture Media, Serum-Free/pharmacology , Fluorescent Antibody Technique , Gene Expression Regulation, Neoplastic/drug effects , Humans , Intracellular Fluid/drug effects , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/metabolism , Nasopharyngeal Neoplasms/pathology , Poly(ADP-ribose) Polymerases/metabolism , Protein Binding , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Transfection
17.
PLoS One ; 8(1): e51459, 2013.
Article in English | MEDLINE | ID: mdl-23326314

ABSTRACT

BACKGROUND: 3T3-L1 cells are widely used to study adipogenesis and insulin response. Their adipogenic potential decreases with time in the culture. Expressing exogenous genes in 3T3-L1 cells can be challenging. This work tries to establish and characterize an alternative model of cultured adipocytes that is easier to work with than the 3T3-L1 cells. METHODOLOGY/PRINCIPAL FINDINGS: INDUCED CELLS WERE IDENTIFIED AS ADIPOCYTES BASED ON THE FOLLOWING THREE CHARACTERISTICS: (1) Accumulation of triglyceride droplets as demonstrated by oil red O stain. (2) Transport rate of 2-deoxyglucose increased after insulin stimulation. (3) Expression of fat specific genes such as Fabp4 (aP2), Slc2a4 (Glut4) and Pparg (PPARγ). Among the cell lines induced under different conditions in this study, only NIH/3T3 cells differentiated into adipocytes after prolonged incubation in 3T3-L1 induction medium containing 20% instead of 10% fetal bovine serum. Rosiglitazone added to the induction medium shortened the incubation period from 14 to 7 days. The PI3K/AKT pathway showed similar changes upon insulin stimulation in these two adipocytes. C/EBPα mRNA was barely detectable in NIH/3T3 adipocytes. NIH/3T3 adipocytes induced in the presence of rosiglitazone showed higher 2-deoxyglucose transport rate after insulin stimulation, expressed less Agt (angiotensinogen) and more PPARγ. Knockdown of C/EBPα using shRNA blocked 3T3-L1 but not NIH/3T3 cell differentiation. Mouse adipose tissues from various anatomical locations showed comparable levels of C/EBPα mRNA. CONCLUSIONS/SIGNIFICANCE: NIH/3T3 cells were capable of differentiating into adipocytes without genetic engineering. They were an adipocyte model that did not require the reciprocal activation between C/EBPα and PPARγ to differentiate. Future studies in the C/EBPα independent pathways leading to insulin responsiveness may reveal new targets to diabetes treatment.


Subject(s)
Adipocytes/drug effects , Adipogenesis/drug effects , CCAAT-Enhancer-Binding Protein-alpha/genetics , Culture Media/pharmacology , 3T3-L1 Cells , Adipocytes/cytology , Adipocytes/metabolism , Adipogenesis/genetics , Animals , Biological Transport/drug effects , Blotting, Western , CCAAT-Enhancer-Binding Protein-alpha/metabolism , Cattle , Cell Differentiation/drug effects , Cell Differentiation/genetics , Culture Media/chemistry , Deoxyglucose/metabolism , Fatty Acid-Binding Proteins/genetics , Fatty Acid-Binding Proteins/metabolism , Gene Expression/drug effects , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Mice , Mice, Inbred C57BL , NIH 3T3 Cells , PPAR gamma/genetics , PPAR gamma/metabolism , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Rosiglitazone , Serum , Thiazolidinediones/pharmacology , Time Factors , Triglycerides/metabolism
18.
Cancer Lett ; 319(2): 203-213, 2012 Jun 28.
Article in English | MEDLINE | ID: mdl-22306340

ABSTRACT

Cisplatin is an anticancer agent that is effective against several types of cancer, including gastric cancer. However, its therapeutic application is limited by its toxicity in normal tissues and complications caused in patients. In this study, we attempted to clarify how triptolide, an active component extracted from the traditional Chinese herbal medicine Tripterygium wilfordii Hook F (TWHF), enhances cisplatin-induced cytotoxicity in gastric cancer SC-M1 cells. After low-dose combined treatments with triptolide and cisplatin, a decrease in viability with a concomitant increase in apoptosis was observed in SC-M1 cells but not in normal cells. Apoptosis induced by the combined treatments was accompanied by loss of mitochondrial membrane potential and release of cytochrome c. Triptolide increased the cisplatin-induced activation of caspase-3 and caspase-9 and the downstream cleavage of PARP in SC-M1 cells. Results of these in vitro experiments indicated that triptolide enhanced cytotoxicity in cisplatin-treated SC-M1 cells and that this effect is mediated by apoptosis through a mitochondrial pathway. Furthermore, using a SCID mouse xenograft model, we demonstrated that the combined treatment completely suppressed tumor growth via down-regulation of proliferating cell nuclear antigen expression without significant side effects. These results suggest that lower concentrations of cisplatin and triptolide used in combination may produce a synergistic anticancer effect that warrants further investigation for its potential clinical applications.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cisplatin/administration & dosage , Diterpenes/administration & dosage , Phenanthrenes/administration & dosage , Stomach Neoplasms/drug therapy , Animals , Apoptosis/drug effects , Cell Line, Tumor , Drug Synergism , Epoxy Compounds/administration & dosage , Humans , Male , Mice , Mice, SCID , Phytotherapy/methods , Plant Extracts , Tripterygium , Xenograft Model Antitumor Assays
19.
Int J Radiat Oncol Biol Phys ; 77(2): 566-74, 2010 Jun 01.
Article in English | MEDLINE | ID: mdl-20457353

ABSTRACT

PURPOSE: Radiotherapy is one of the best choices for cancer treatment. However, various tumor cells exhibit resistance to irradiation-induced apoptosis. The development of new strategies to trigger cancer cell death besides apoptosis is necessary. This study investigated the role of securin in radiation-induced apoptosis and senescence in human cancer cells. METHODS AND MATERIALS: Cell survival was determined using clonogenic assays. Western blot analysis was used to analyze levels of securin, caspase-3, PARP, p53, p21, Rb, gamma-H2AX, and phospho-Chk2. Senescent cells were analyzed using a beta-galactosidase staining assay. A securin-expressed vector (pcDNA-securin) was stably transfected into securin-null HCT116 cells. Securin gene knockdown was performed by small interfering RNA and small hairpin RNA in HCT116 and MDA-MB-231 cells, respectively. RESULTS: Radiation was found to induce apoptosis in securin wild type HCT116 cells but induced senescence in securin-null cells. Restoration of securin reduced senescence and increased cell survival in securin-null HCT116 cells after irradiation. Radiation-induced gamma-H2AX and Chk2 phosphorylation were induced transiently in securin-wild-type cells but exhibited sustained activation in securin-null cells. Securin gene knockdown switches irradiation-induced apoptosis to senescence in both HCT116 p53-null and MDA-MB-231 cells. CONCLUSIONS: Our results demonstrated that the level of securin expression plays a determining role in the radiosensitivity and fate of cells. Depletion of securin impairs DNA repair after irradiation, increasing DNA damage and promoting senescence in the residual surviving cells regardless of functional p53 expression. The knockdown of securin may contribute to a novel radiotherapy protocol for the treatment of human cancer cells that are resistant to irradiation.


Subject(s)
Apoptosis/physiology , Cellular Senescence/physiology , Neoplasm Proteins/metabolism , Radiation Tolerance/physiology , Tumor Suppressor Protein p53/metabolism , Apoptosis/radiation effects , Blotting, Western , Caspase 3/metabolism , Cell Line, Tumor/radiation effects , Cellular Senescence/radiation effects , Checkpoint Kinase 2 , Colony-Forming Units Assay/methods , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA Repair , Gene Knockdown Techniques/methods , HCT116 Cells/radiation effects , Histones/metabolism , Humans , Neoplasm Proteins/deficiency , Neoplasm Proteins/genetics , Phosphorylation , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/metabolism , Protein Serine-Threonine Kinases/metabolism , Retinoblastoma Protein/metabolism , Securin
20.
Head Neck ; 32(7): 877-85, 2010 Jul.
Article in English | MEDLINE | ID: mdl-19908316

ABSTRACT

BACKGROUND: Prostate apoptosis response-4 (Par-4) augments apoptosis in various tumors, either during apoptotic insult or by ectopic overexpression. However, investigation of Par-4 expression in nasopharyngeal carcinoma (NPC) is lacking. METHODS: Specimens from patients with NPC, hypopharyngeal carcinoma (HPC), or oral cavity cancer were examined for Par-4 expression using immunohistochemistry. NPC cell proliferation and apoptosis were analyzed using immunohistochemical staining for Ki67, B-cell lymphoma 2 (Bcl-2), and in situ terminal deoxyribonucleotidyl transferase (TdT)-mediated biotin-16-dUTP nick end-labeling (TUNEL) assay, respectively. RESULTS: Par-4 was ubiquitously expressed in NPC biopsies (96.2%, 25/26) and was significantly higher than in HPC (47.6%, 50/105, p < .0001) and oral cavity cancers (38.7%, 12/31, p < .0001). Remarkably, apoptosis of NPC cells was absent and Par-4 expression was associated with obvious expression of Bcl-2 and Ki67 in all patients tested with NPC. CONCLUSIONS: Immunohistochemistry results showed widespread expression of Par-4 in NPC and revealed sustainable proliferation of NPC cells regardless of Par-4 expression.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Carcinoma/metabolism , Carcinoma/pathology , Nasopharyngeal Neoplasms/metabolism , Nasopharyngeal Neoplasms/pathology , Apoptosis , Cell Proliferation , Cohort Studies , Female , Humans , Hypopharyngeal Neoplasms/metabolism , Hypopharyngeal Neoplasms/pathology , Ki-67 Antigen/metabolism , Male , Mouth Neoplasms/metabolism , Mouth Neoplasms/pathology , Proto-Oncogene Proteins c-bcl-2/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL