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










Publication year range
1.
J Pharm Biomed Anal ; 186: 113300, 2020 Jul 15.
Article in English | MEDLINE | ID: mdl-32413824

ABSTRACT

Cancer metastasis is the major cause of death in pancreatic cancer. We have established a pair of pancreatic ductal adenocarcinoma cell line, PANC1 and invasive PANC1-I5, as a model system toinvestigate the metastatic mechanism as well as potential therapeutic targets in pancreatic cancer. We used proteomic analysis based on two-dimensional differential gel electrophoresis (2D-DIGE) and matrix-assisted laser desorption ionization time-of-flight mass spectrometry (MALDI-TOF MS) to examine the global protein expression alterations between PANC1 and PANC1-I5. Proteomic study revealed that 88 proteins are differentially expressed between PANC1-I5 and PANC1 cells, and further functional evaluations through protein expression validation, gene knockout, migration and invasion analysis revealed that galectin-1 is one of the potential players in modulating pancreatic cancer metastasis. To conclude, we have identified numerous proteins might be associated with pancreatic cancer invasiveness in the pancreatic cancer model.


Subject(s)
Carcinoma, Pancreatic Ductal/pathology , Galectin 1/metabolism , Pancreatic Neoplasms/pathology , Proteomics , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Biomarkers, Tumor/metabolism , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Cell Line, Tumor , Cell Movement , Gene Expression Regulation, Neoplastic , Gene Knockout Techniques , Humans , Neoplasm Invasiveness , Neoplasm Metastasis , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Two-Dimensional Difference Gel Electrophoresis
2.
Onco Targets Ther ; 12: 6119-6131, 2019.
Article in English | MEDLINE | ID: mdl-31496721

ABSTRACT

BACKGROUND: Kurarinone, a prenylated flavonone isolated from the roots of Sophora flavescens, is known to be cytotoxic against many human cancer cells but not human small cell lung carcinoma (SCLC) yet. Also, the exact molecular mechanism of kurarinone for induction cytotoxicity remains unknown. MATERIAL AND METHODS: We investigated the effects of kurarinone on cell proliferation, apoptosis, and migration in H1688 SCLC cells. Cell viability was determined by the MTT assay. Apoptotic indices such as cell cycle, mitochondrial membrane potential, cytochrome c release, caspase activity, and death receptors were evaluated by flow cytometry. Transwell migration and invasion assays were also included. RESULTS: Our results indicated that kurarinone significantly decreased H1688 cell viability and induced the accumulation of sub-G1 fractions by activating caspase-3, -9, and PARP cleavage accompanied by the elevated release of cytochrome c and mitochondrial dysfunction in H1688 cells. Additionally, kurarinone promoted Fas and TRAIL receptor-1 and -2 expression via the caspase-8/Bid pathway, suggesting that kurarinone triggered apoptosis via the mitochondria-mediated and receptor-mediated apoptotic pathways. We also observed that kurarinone repressed migration and invasion capabilities of SCLC cells by suppressing the expression of epithelial-mesenchymal transition-related proteins and matrix metalloproteinases. CONCLUSION: Our findings provided evidence that kurarinone can induce apoptosis in SCLC cells via multiple mechanisms and delayed the cell migration and invasion of SCLC cells.

3.
Ci Ji Yi Xue Za Zhi ; 31(2): 81-85, 2019.
Article in English | MEDLINE | ID: mdl-31007486

ABSTRACT

OBJECTIVE: Naringenin, a flavonoid found in citrus fruits, has notably diverse pharmacological properties. In the present study, we investigated the antinociceptive and anti-inflammatory effects of naringenin. MATERIALS AND METHODS: The antinociceptive effects were evaluated using hot-plate, acetic acid-induced writhing, and tail-flick assays in mice and rats. The anti-inflammatory effects were examined by a carrageenan-induced paw edema test in rats. RESULTS: Naringenin (100 or 200 mg/kg, oral administration) significantly delayed the reaction time of mice to thermal stimulation generated by a hot plate and a tail-flick unit and reduced the acetic acid-induced writhing response in mice. In addition, naringenin significantly decreased paw edema induced by carrageenan in rats, showing its anti-inflammatory effect. CONCLUSION: Our results show that naringenin has therapeutic potential with antinociceptive and anti-inflammatory properties and can further be exploited for the development of drugs for pain and inflammatory-related diseases.

4.
Mol Med Rep ; 16(5): 6992-7000, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28901434

ABSTRACT

The dendritic cell (DC) maturation process is essential for the development of T cell responses and immune tolerance. Accordingly, DCs are considered a major target in the development of immunomodulating agents. In the present study, the effect of sinulariolide, an active compound isolated from the cultured soft coral Sinularia flexibilis, on lipopolysaccharide (LPS)­induced murine bone marrow­derived DCs was evaluated. The different phenotypes, cytokine secretion and the mix­lymphocyte reaction of DCs were detected using flow cytometry and ELISA. The experimental results revealed that the phenotypic and functional maturation of DCs stimulated by LPS were markedly reduced by sinulariolide in a concentration­dependent manner, including the expression of co­stimulatory molecules (CD40, CD80 and CD86). In addition, sinulariolide reduced the release of tumor necrosis factor­α, interleukin (IL)­6, IL­12 and nitric oxide from the LPS­activated DCs, decreased their abilities to stimulate allogeneic T cell proliferation, and inhibited LPS­induced nuclear factor­κB pathways. These findings offer novel insight into the immunopharmacological function of sinulariolide and its effects on DCs.


Subject(s)
Cell Proliferation/drug effects , Diterpenes/pharmacology , Lipopolysaccharides/toxicity , Lymphocyte Activation/drug effects , Animals , B7-1 Antigen/metabolism , B7-2 Antigen/metabolism , Bone Marrow Cells/cytology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD40 Antigens/metabolism , Cell Survival/drug effects , Cells, Cultured , Dendritic Cells/cytology , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Interleukin-12/analysis , Interleukin-6/analysis , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Nitric Oxide/analysis , Phenotype
5.
Mar Drugs ; 15(7)2017 Jul 01.
Article in English | MEDLINE | ID: mdl-28671570

ABSTRACT

24-methyl-cholesta-5,24(28)-diene-3ß,19-diol-7ß-monoacetate (MeCDDA) is a natural steroid compound isolated from a wild-type soft coral (Nephthea erecta). The present study aimed to investigate the anti-small cell lung cancer (SCLC) effects of MeCDDA in vitro and in vivo, as well as to elucidate its underlying mechanism. Our results indicated that H1688 and H146 cells show relevant sensitivity to MeCDDA, and the exposure to MeCDDA in SCLC cells caused dose-dependent growth inhibitory responses. In addition, MeCDDA treatment promoted cell apoptosis and increased the activities of caspases in H1688 cells, reducing the mitochondrial membrane potential and stimulating the release of cytochrome c into the cytosol. Along with the increase in Bax expression and reduction in Bcl-2, the MeCDDA treatment also significantly decreased Akt and mTOR phosphorylation. Finally, MeCDDA treatment in the mouse xenograft model of H1688 cells exhibited significant inhibition of tumor growth, corroborating MeCDDA as a potential pre-clinical candidate for the treatment of SCLC. Overall, our results demonstrate that the cytotoxic effects of MeCDDA towards H1688 and H146 cells, possibly through the activation of the mitochondrial apoptotic pathway and inhibition of the PI3K/Akt/mTOR pathway, merit further studies for its possible clinical application in chemotherapy.


Subject(s)
Anthozoa/chemistry , Antineoplastic Agents/pharmacology , Small Cell Lung Carcinoma/drug therapy , Steroids/pharmacology , Animals , Anthozoa/metabolism , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Apoptosis/drug effects , Caspases/genetics , Caspases/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Steroids/chemistry , Steroids/metabolism
6.
BMC Complement Altern Med ; 17(1): 62, 2017 Jan 19.
Article in English | MEDLINE | ID: mdl-28103869

ABSTRACT

BACKGROUND: Sinularin isolated from the cultured soft coral Sinularia flexibilis has been reported to exert potent cytotoxic effects against particular types of cancer. This study was carried out to investigate the cytotoxic effects in sinularin-treated human hepatocellular carcinoma cells, HepG2, and to subsequently explore the underlying molecular mechanisms. METHODS: TheMTT (3-[4,5-dimethylthiazol-2-yl]-2, 5-diphenyl- tetrazolium bromide) method was used to evaluate the cytotoxicity of sinularin on HepG2 and Hep3B cell lines. Furthermore, the cell cycle distribution assay, apoptosis assay, and western blot analysis in vitro were used to explore the possible mechanisms of action. RESULTS: From the results of our study, cell viability was obviously inhibited by sinularin in a dose-dependent manner. In addition, our results suggested that sinularin triggered DNA damage and subsequently induced cell cycle G2/M arrest associated with up-regulation of p-ATM (Ser(1981)), p-Chk2 (Tyr(68)), p-cdc2 (Tyr(15)), and p53 coupled with increased expression of downstream proteins p21 and down-regulation of p-cdc25 (Ser(216)). Moreover, the results of the apoptosis assay and western blot analysis indicated that the cytotoxic activity could be related to mitochondrial apoptosis, characterized by decrease of Bcl-2 expression, disruption of mitochondrial membrane potential, and sequential activation of caspases and Poly (ADP-ribose) polymerase (PARP). CONCLUSIONS: This study reveals for the first time the anti-HCC activities of sinularin, the active compound isolated from the cultured soft coral Sinularia flexibilis. We believe that our results warrant further evaluation of sinularin as a new anti-HCC chemotherapeutic agent.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Hepatocellular/drug therapy , DNA Damage/drug effects , Diterpenes/pharmacology , Heterocyclic Compounds, 3-Ring/pharmacology , Liver Neoplasms/drug therapy , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Division/drug effects , Cell Survival/drug effects , G2 Phase/drug effects , Hep G2 Cells , Humans , Membrane Potential, Mitochondrial/drug effects
8.
Oncotarget ; 7(13): 17144-61, 2016 03 29.
Article in English | MEDLINE | ID: mdl-26958809

ABSTRACT

The oral cancer cell line OC3-I5 with a highly invasive ability was selected and derived from an established OSCC line OC3. In this study, we demonstrated that matrix metalloproteinases protein MMP-13 was up-regulated in OC3-I5 than in OC3 cells. We also observed that expression of epithelial-mesenchymal transition (EMT) markers including Twist, p-Src, Snail1, SIP1, JAM-A, and vinculin were increased in OC3-I5 compared to OC3 cells, whereas E-cadherin expression was decreased in the OC3-I5 cells. Using siMMP-13 knockdown techniques, we showed that siMMP-13 not only reduced the invasion and migration, but also the adhesion abilities of oral cancer cells. In support of the role of MMP-13 in metastasis, we used MMP-13 expressing plasmid-transfected 293T cells to enhance MMP-13 expression in the OC3 cells, transplanting the MMP-13 over expressing OC3 cells into nude mice led to enhanced lung metastasis. In summary, our findings show that MMP-13 promotes invasion and metastasis in oral cancer cells, suggesting altered expression of MMP-13 may be utilized to impede the process of metastasis.

9.
Int Immunopharmacol ; 33: 70-82, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26874324

ABSTRACT

Psoriasis is a chronic autoimmune disease of undefined etiology that involves dysregulated interplay between immune cells and keratinocytes. Acarbose was found to decrease inflammatory parameters in diabetic patients in addition to its anti-diabetic effects. Here, we report that imiquimod (IMQ)-induced epidermal hyperplasia and psoriasis like-inflammation were significantly inhibited by acarbose treatment. Real-time PCR showed that mRNA levels of the cytokines TNF-α, IL-6, IL-1ß IL-17A, and IL-22 in skin were also decreased significantly by acarbose. In addition, we found that acarbose reduced infiltration of CD3(+) T cells and GR-1(+) neutrophils in lesional skin and also reduced the percentage of IL-17-producing CD4(+) T cells (Th17) and IL-17- and IL-22-producing γδ T cells in the spleen. In contrast, acarbose increased the frequency of IL-10-producing CD4(+) regulator Tr1 T cells in the spleen and small intestine. These results indicate that oral administration of acarbose can attenuate the severity of imiquimod-induced psoriasis with local and systemic anti-inflammatory and immune modulation effects, thus suggesting that acarbose is an effective therapeutic strategy for psoriasis regulation.


Subject(s)
Acarbose/therapeutic use , Dermatitis, Contact/drug therapy , Neutrophils/drug effects , Psoriasis/drug therapy , Skin/drug effects , T-Lymphocytes, Regulatory/drug effects , Th17 Cells/drug effects , Acarbose/pharmacology , Administration, Oral , Aminoquinolines , Animals , Cell Movement/drug effects , Cytokines/metabolism , Disease Models, Animal , Humans , Imiquimod , Mice , Mice, Inbred BALB C , Neutrophils/immunology , Psoriasis/chemically induced , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Skin/immunology , Skin/pathology , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology
10.
Cancer Biol Ther ; 17(1): 48-64, 2016.
Article in English | MEDLINE | ID: mdl-26569090

ABSTRACT

Tangeretin, a major phytochemicals in tangerine peels--an important Chinese herb, has been found to have anti-carcinogenic properties. To improve bioavailability and increase potency of tangeretin, its derivative, 5-acetyloxy-6,7,8,4'-tetramethoxyflavone (5-AcTMF), has been synthesized and shown potent inhibition of proliferation activity against human breast and leukemia cancer cell lines. In this study, we have further investigated the anticancer effects of 5-AcTMF on CL1-5 non-small cell lung cancer cells (NSCLC) both in vitro and in vivo and demonstrated that 5-AcTMF effectively inhibited cancer cell proliferation, induced G2/M-phase arrest associated with cdc2 and CDC25c and increased in the apoptotic cells associated with caspase activation, down regulation of Bcl-2, XIAP and Survivn, inducing release of cytochrome c into the cytosol and disruption of mitochondrial membrane potential. We also found that 5-AcTMF treatment of CL1-5 activated autophagy, indicated by triggered autophagosome formation and increased LC3-II levels and formation of LC3 puncta. Moreover, we also found that 5-AcTMF lowered phophoatidylinositol 3-kinase/AKT/mTOR signaling pathway. Over-expression of AKT by AKT cDNA transfection decreased 5-AcTMF mediated apoptosis and autophagy, supporting the induction of apoptosis and autophagy by inhibition of AKT pathway. In an animal study, 5-AcTMF effectively delayed tumor growth in a nude mouse model of CL1-5 xenografts without observed adverse effect. Immunohistochemistry Analysis indicated that 5-AcTMF induced CL1-5 cell apoptosis and autophagy in vivo. Taken together, these data demonstrate that 5-AcTMF is a novel small molecule agent that can inhibit NSCLC cell proliferation, and induce G(2)/M phase arrest and via the mitochondrial apoptotic pathway and autophagy.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Cell Cycle Checkpoints/drug effects , Flavones/administration & dosage , Animals , Apoptosis/drug effects , Autophagy/drug effects , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Flavones/chemical synthesis , Flavones/chemistry , G2 Phase Cell Cycle Checkpoints/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Membrane Potential, Mitochondrial/drug effects , Mice , Proto-Oncogene Proteins c-akt/biosynthesis , Xenograft Model Antitumor Assays
11.
J Proteome Res ; 14(11): 4907-20, 2015 Nov 06.
Article in English | MEDLINE | ID: mdl-26452990

ABSTRACT

Drug resistance is one of the major causes of cancer chemotherapy failure. In the current study, we used a pair of lung adenocarcinoma cell lines, A549 and the pemetrexed-resistant A549/PEM cells, as a model to monitor resistance-dependent cellular responses and identify potential therapeutic targets. By means of 2D differential gel electrophoresis (2D-DIGE) and matrix-assisted laser desorption ionization time-of-flight mass spectrometry (MALDI-TOF MS), we investigated the global protein expression alterations induced by pemetrexed treatment and resistance. The proteomic result revealed that pemetrexed exposure obviously altered the expression of 81 proteins in the A549 cells, whereas no significant response was observed in the similarly treated A549/PEM cells, hence implying an association between these proteins and the drug-specific response. Moreover, 72 proteins including flavin reductase and calreticulin demonstrated differential expression between the A549 and A549/PEM cells, indicating baseline resistance. Additional tests employed siRNA silencing, protein overexpression, cell viability analysis, and analysis of apoptosis to examine and confirm the potency of flavin reductase and calreticulin proteins in the development of pemetrexed resistance. In summary, by using a proteomic approach, we identified numerous proteins, including flavin reductase and calreticulin, involved in pemetrexed drug resistance-developing mechanisms. Our results provide useful diagnostic markers and therapeutic candidates for pemetrexed-resistant lung cancer treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Calreticulin/isolation & purification , FMN Reductase/isolation & purification , Gene Expression Regulation, Neoplastic , Neoplasm Proteins/genetics , Pemetrexed/pharmacology , Proteome/isolation & purification , Adenocarcinoma/drug therapy , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Apoptosis/drug effects , Calreticulin/genetics , Calreticulin/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Drug Resistance, Neoplasm/genetics , Electrophoresis, Gel, Two-Dimensional , FMN Reductase/genetics , FMN Reductase/metabolism , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Proteome/genetics , Proteome/metabolism , Proteomics/methods , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
12.
Toxicol Appl Pharmacol ; 288(3): 359-73, 2015 Nov 01.
Article in English | MEDLINE | ID: mdl-26298006

ABSTRACT

Gefitinib is the first-line chemotherapeutic drug for treating non-small cell lung cancer (NSCLC), which comprises nearly 85% of all lung cancer cases worldwide. However, most patients eventually develop drug resistance after 12-18 months of treatment. Hence, investigating the drug resistance mechanism and resistance-associated biomarkers is necessary. Two lung adenocarcinoma cell lines, PC9 and gefitinib-resistant PC9/Gef, were established for examining resistance mechanisms and identifying potential therapeutic targets. Two-dimensional differential gel electrophoresis and matrix-assisted laser desorption ionization time-of-flight mass spectrometry were used for examining global protein expression changes between PC9 and PC9/Gef. The results revealed that 164 identified proteins were associated with the formation of gefitinib resistance in PC9 cells. Additional studies using RNA interference showed that progesterone receptor membrane component 1 and pericentrin proteins have major roles in gefitinib resistance. In conclusion, the proteomic approach enabled identifying of numerous proteins involved in gefitinib resistance. The results provide useful diagnostic markers and therapeutic candidates for treating gefitinib-resistant NSCLC.


Subject(s)
Adenocarcinoma/pathology , Carcinoma, Non-Small-Cell Lung/pathology , Drug Resistance, Neoplasm , Lung Neoplasms/drug therapy , Quinazolines/pharmacology , Adenocarcinoma/metabolism , Adenocarcinoma of Lung , Antigens/genetics , Antigens/metabolism , Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor/drug effects , Cell Survival/drug effects , ErbB Receptors/genetics , ErbB Receptors/metabolism , Gefitinib , Gene Silencing , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Proteomics/methods , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, Progesterone/genetics , Receptors, Progesterone/metabolism , Signal Transduction , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
13.
J Nutr Biochem ; 26(5): 484-504, 2015 May.
Article in English | MEDLINE | ID: mdl-25765513

ABSTRACT

5-Demethylnobiletin is a hydroxylated polymethoxyflavone found in citrus plants that shows antiproliferative activities in several cancer cell lines. In this study, we investigated the effects and underlying molecular mechanisms of 5-demethylnobiletin on inhibition of cell growth, apoptosis, cell cycle and autophagy in A549 and CL1-5 lung cancer cells. The results of the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay suggested that 5-demethylnobiletin inhibited cell growth in a dose- and time-dependent manner. Flow cytometry results suggested that 5-demethylnobiletin inhibited proliferation in lung cancer cells by inducing G2/M cell cycle phase arrest but predominantly not through apoptosis. Western blot results illustrated that the blockade of the cell cycle was associated with reduced levels of cdc25 and cdc2. Notably, our results indicated that 5-demethylnobiletin induced significant abnormal microtubule dynamics in A549 and CL1-5 cells, a novel finding. Studies conducted with isolated tubulin and docking models suggest that 5-demethylnobiletin promoted the polymerization of microtubules and bound to the taxol site. Additionally, 5-demethylnobiletin might also induce autophagy via activation of the JNK signaling pathway in A549 and CL1-5 cells. Pretreatment of the cells with the autophagy inhibitor 3-methyladenine significantly potentiated 5-demethylnobiletin-induced apoptosis, suggesting that 5-demethylnobiletin-induced autophagy mitigated cell apoptosis. Further investigation revealed that 5-demethylnobiletin inhibition of CL1-5 lung cancer cell growth was reproducible in a nude mouse model. Taken together, these studies suggest that 5-demethylnobiletin has anti-lung cancer efficacy both in vitro and in vivo possibly through induction of G2/M arrest, autophagy and apoptosis.


Subject(s)
Autophagy/drug effects , Cell Division/drug effects , Flavones/pharmacology , G2 Phase/drug effects , Lung Neoplasms/pathology , Tubulin/biosynthesis , Cell Line, Tumor , DNA Damage , Flavones/isolation & purification , Humans , Lung Neoplasms/metabolism , Polymerization , Tubulin/chemistry
14.
Asian Pac J Cancer Prev ; 15(18): 7995-8001, 2014.
Article in English | MEDLINE | ID: mdl-25292102

ABSTRACT

Interferon-gamma (IFN-γ) has been used to treat various malignant tumors. However, the molecular mechanisms underlying the direct anti-proliferative activity of IFN-γ are poorly understood. In the present study, we examined the in vitro antitumor activity of IFN-γ on two human non-small-cell lung carcinoma (NSCLC) cell lines, H322M and H226. Our findings indicated that IFN-γ treatment caused a time-dependent reduction in cell viability and induced apoptosis through a FADD-mediated caspase-8/tBid/mitochondria-dependent pathway in both cell lines. Notably, we also postulated that IFN-γ increased indoleamine 2,3-dioxygenase (IDO) expression and enzymatic activity in H322M and H226 cells. In addition, inhibition of IDO activity by the IDO inhibitor 1-MT or tryptophan significantly reduced IFN-γ-induced apoptosis and death receptor 5 (DR5) expression, which suggests that IDO enzymatic activity plays an important role in the anti-NSCLC cancer effect of IFN-γ. These results provide new mechanistic insights into interferon-γ antitumor activity and further support IFN-γ as a potential therapeutic adjuvant for the treatment of NCSLC.


Subject(s)
Apoptosis/drug effects , Carcinoma, Non-Small-Cell Lung/pathology , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Interferon-gamma/pharmacology , Lung Neoplasms/pathology , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Adenocarcinoma/drug therapy , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Antiviral Agents/pharmacology , Blotting, Western , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Cycle/drug effects , Cell Proliferation/drug effects , Cytochromes c/metabolism , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Membrane Potential, Mitochondrial/drug effects , Tumor Cells, Cultured
15.
Article in English | MEDLINE | ID: mdl-23476680

ABSTRACT

Acute lymphoblastic leukemia (ALL) accounts for approximately 75% of childhood leukemia, and chemotherapy remains the mainstay therapy. Baicalein is an active flavonoid used in traditional Chinese medicine and has recently been found to have anticancer, anti-inflammatory, and antiallergic properties. This study aims to investigate the molecular apoptotic mechanisms of baicalein in CCRF-CEM leukemic cells and to evaluate the combined therapeutic efficacy of baicalein with several commonly used chemotherapeutic drugs in CCRF-CEM cells. Our results demonstrate that baicalein induces mitochondria-dependent cleavage of caspases-9 and -3 and PARP with concomitant decreases in IAP family proteins, survivin, and XIAP. Furthermore, our results present for the first time that baicalein triggers a convergence of the intrinsic and extrinsic apoptotic pathways via the death receptor-caspase 8-tBid signaling cascade in CCRF-CEM cells. In addition, we also present for the first time that the combination of baicalein and vincristine results in a synergistic therapeutic efficacy. Overall, this combination strategy is recommended for future clinical trials in the treatment of pediatric leukemia owing to baicalein's beneficial effects in alleviating the vomiting, nausea, and skin rashes caused by chemotherapy.

SELECTION OF CITATIONS
SEARCH DETAIL
...