Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Acta Pharmacol Sin ; 44(2): 434-445, 2023 Feb.
Article in English | MEDLINE | ID: mdl-35945312

ABSTRACT

Interleukin (IL)-1ß is a culprit of adipose tissue inflammation, which in turn causes systematic inflammation and insulin resistance in obese individuals. IL-1ß is mainly produced in monocytes and macrophages and marginally in adipocytes, through cleavage of the inactive pro-IL-1ß precursor by caspase-1, which is activated via the NLRP3 inflammasome complex. The nuclear factor-κB (NF-κB) transcription factor is the master regulator of inflammatory responses. Brindle berry (Garcinia cambogia) has been widely used as health products for treating obesity and related metabolic disorders, but its active principles remain unclear. We previously found a series of polyisoprenylated benzophenones from brindle berry with anti-inflammatory activities. In this study we investigated whether 14-deoxygarcinol (DOG), a major polyisoprenylated benzophenone from brindle berry, alleviated adipose tissue inflammation and insulin sensitivity in high-fat diet fed mice. The mice were administered DOG (2.5, 5 mg · kg-1 · d-1, i.p.) for 4 weeks. We showed that DOG injection dose-dependently improved insulin resistance and hyperlipidemia, but not adiposity in high-fat diet-fed mice. We found that DOG injection significantly alleviated adipose tissue inflammation via preventing macrophage infiltration and pro-inflammatory polarization of macrophages, and adipose tissue fibrosis via reducing the abnormal deposition of extracellular matrix. In LPS plus nigericin-stimulated THP-1 macrophages, DOG (1.25, 2.5, 5 µM) dose-dependently suppressed the activation of NLRP3 inflammasome and NF-κB signaling pathway. We demonstrated that DOG bound to and activated the deacetylase Sirtuin 2, which in turn deacetylated and inactivated NLRP3 inflammasome to reduce IL-1ß secretion. Moreover, DOG (1.25, 2.5, 5 µM) dose-dependently mitigated inflammatory responses in macrophage conditioned media-treated adipocytes and suppressed macrophage migration toward adipocytes. Taken together, DOG might be a drug candidate to treat metabolic disorders through modulation of adipose tissue remodeling.


Subject(s)
Insulin Resistance , NF-kappa B , Animals , Mice , Adipose Tissue/metabolism , Diet, High-Fat/adverse effects , Inflammasomes/metabolism , Inflammation/drug therapy , Inflammation/metabolism , Insulin/metabolism , Insulin Resistance/physiology , Mice, Obese , NF-kappa B/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Obesity/drug therapy , Obesity/metabolism , Sirtuin 2/metabolism
2.
Acta Pharmacol Sin ; 43(7): 1780-1792, 2022 Jul.
Article in English | MEDLINE | ID: mdl-34789920

ABSTRACT

Interleukin-1ß (IL-1ß), a key pro-inflammatory cytokine, is majorly produced by macrophages through NOD-, LRR-, and pyrin domain-containing protein 3 (NLRP3) inflammasome, which has been identified as the culprit to deteriorate the inflammatory crosstalk between macrophages and adipocytes. Ainsliadimer C (AC) is a disesquiterpenoid isolated from Ainsliaea macrocephala. In the current study, we investigated the effects of AC on adipose tissue inflammation in co-culture of macrophages and adipocytes in vitro as well as in LPS-treated mice in vivo. We showed that AC (20-80 µM) dose-dependently inhibited the secretion of IL-1ß from LPS plus ATP-stimulated THP-1 macrophages by inhibiting the activation of NLRP3 inflammasome. Furthermore, we found that AC treatment activated NAD+-dependent deacetylase Sirtuin 1 (SIRT1), resulting in reduced acetylation level of NLRP3. Molecular modeling analysis revealed that binding of AC to sirtuin-activating compound-binding domain increased the affinity of the substrate to the catalytic domain of SIRT1. Moreover, AC (80 µM) significantly attenuated macrophage-conditioned medium-induced inflammatory responses in 3T3-L1 adipocytes. In LPS-induced acute inflammatory mice, administration of AC (20, 60 mg·kg-1·d-1, ip) for 5 days significantly suppressed the pro-inflammatory cytokine levels in serum and epididymal white adipose tissue (eWAT), attenuated macrophage infiltration into eWAT, and mitigated adipose tissue inflammation. The beneficial effects of AC were blocked by co-administration of a selective SIRT1 inhibitor EX-527 (10 mg·kg-1·d-1). Taken together, AC suppresses NLRP3-mediated IL-1ß secretion through activating SIRT1, leading to attenuated inflammation in macrophages and adipose tissue, which might be a candidate to treat obesity-associated metabolic diseases.


Subject(s)
Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Adipose Tissue/metabolism , Animals , Cytokines/metabolism , Inflammasomes/metabolism , Inflammation/drug therapy , Inflammation/metabolism , Interleukin-1beta/metabolism , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred NOD , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Sirtuin 1/metabolism
3.
Chin J Nat Med ; 18(7): 517-525, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32616192

ABSTRACT

Nagilactone E (NLE), a natural product with anticancer activities, is isolated from Podocarpus nagi. In this study, we reported that NLE increased programmed death ligand 1 (PD-L1) expressions at both protein and mRNA levels in human lung cancer cells, and enhanced its localization on the cell membrane. Mechanistically, NLE increased the phosphorylation and expression of c-Jun, and promoted the localization of c-Jun in the nucleus, while silencing of c-Jun by small interfering RNA (siRNA) reduced NLE-induced PD-L1. Further study showed that NLE activated the c-Jun N-terminal kinases (JNK), the upstream of c-Jun, and its inhibitor SP600125 reversed the NLE-increased PD-L1. Moreover, NLE-induced PD-L1 increased the binding intensity of PD-1 on the cell surface. In summary, NLE upregulates the expression of PD-L1 in lung cancer cells through the activation of JNK-c-Jun axis, which has the potential to combine with the PD-1/PD-L1 antibody therapies in lung cancer.


Subject(s)
Antineoplastic Agents/pharmacology , B7-H1 Antigen/metabolism , Diterpenes/pharmacology , JNK Mitogen-Activated Protein Kinases/metabolism , Lactones/pharmacology , Antineoplastic Agents/chemistry , Cell Line, Tumor , Diterpenes/chemistry , Humans , Lactones/chemistry , Molecular Structure
4.
Toxicol Appl Pharmacol ; 389: 114882, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31953203

ABSTRACT

Pulmonary fibrosis is a prototypic chronic progressive lung disease with high morbidity and mortality worldwide. Novel effective therapeutic agents are urgently needed owing to the limited treatment options in clinic. Herein, nagilactone D (NLD), a natural dinorditerpenoid obtained from Podocarpus nagi, was found to suppress transforming growth factor-ß1 (TGF-ß1)-mediated fibrotic process in vitro and bleomycin (BLM)-induced pulmonary fibrosis in vivo. NLD attenuated TGF-ß1-induced expression of fibrotic markers including type I and III collagen, fibronectin, α-SMA, and CTGF in human pulmonary fibroblasts (WI-38 VA-13 and HLF-1 cells). Mechanism study indicated that NLD suppressed TGF-ß1-induced up-regulation of TßR I, and Smad2 phosphorylation, nuclear translocation, and transcriptional activation. Moreover, NLD ameliorated BLM-induced histopathological abnormalities in the lungs of experimental fibrotic mice, suppressed synthesis of relative fibrotic markers and fibroblast-to-myofibroblast transition, as well as BLM-induced up-regulation of TßR I expression and Smad signaling in mouse lungs. These data collectively support NLD to be a potential therapeutic agent for pulmonary fibrosis.


Subject(s)
Diterpenes/pharmacology , Fibroblasts/drug effects , Pulmonary Fibrosis/drug therapy , Smad2 Protein/metabolism , Terpenes/pharmacology , Transforming Growth Factor beta1/metabolism , Animals , Biomarkers/metabolism , Bleomycin/pharmacology , Cells, Cultured , Female , Fibroblasts/metabolism , Humans , Lung/drug effects , Lung/metabolism , Mice , Mice, Inbred C57BL , Myofibroblasts/drug effects , Myofibroblasts/metabolism , Pulmonary Fibrosis/metabolism , Receptor, Transforming Growth Factor-beta Type I/metabolism , Signal Transduction/drug effects
5.
Phytomedicine ; 52: 32-39, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30599910

ABSTRACT

BACKGROUND: Non-small cell lung cancer (NSCLC) is one of the leading causes of cancer-related death around the world. Epithelial-mesenchymal transition (EMT) has been documented to increase motility and invasiveness of cancer cells, which promotes cancer metastasis. PURPOSE: This study aims to investigate the inhibitory effects and mechanisms of the dinorditerpenoids and norditerpenoids isolated from the seeds of Podocarpus nagi against transforming growth factor (TGF)-ß1-induced EMT. METHODS: A series of dinorditerpenoids and norditerpenoids were isolated from the seeds of P. nagi. Western blot and quantitative real-time PCR assays were performed to determine the expression levels of relative proteins and mRNA, along with immunofluorescence, Smad-binding element (SBE)-luciferase and chromatin immunoprecipitation (ChIP) assays for the mechanism study. Transwell assays were conducted to determine the effect of the compounds on cell migration and invasion. RESULTS: Nagilactone E (NLE) showed the superior inhibitory effect against TGF-ß1-induced EMT. NLE treatment dramatically inhibited TGF-ß1-induced expression of EMT markers in A549 cells. Mechanism study indicated that NLE markedly suppressed TGF-ß1-induced Smad2 and Smad3 activation and nuclear translocation. SBE-luciferase and ChIP assays showed that NLE inhibited the combining of Smad3 to SBE in the promoters of the cell signaling factors. NLE co-treatment attenuated TGF-ß1-induced up-regulation of the protein and mRNA levels of TGF-ß receptor TßRI. Furthermore, NLE inhibited TGF-ß1-stimulated cell migration and invasion, as well as up-regulation of the key signaling proteins related with migration and invasion. CONCLUSION: NLE inhibited TGF-ß/Smad signaling pathway, thereafter suppressed TGF-ß1-induced EMT, migration and invasion in NSCLC A549 cells.


Subject(s)
Carcinoma, Non-Small-Cell Lung/pathology , Diterpenes/pharmacology , Epithelial-Mesenchymal Transition/drug effects , Lung Neoplasms/pathology , Transforming Growth Factor beta1/pharmacology , A549 Cells , Cell Movement/drug effects , Ferns/chemistry , Humans , Neoplasm Invasiveness , Seeds/chemistry , Signal Transduction/drug effects , Smad2 Protein/metabolism , Smad3 Protein/metabolism
6.
Chin Med ; 13: 34, 2018.
Article in English | MEDLINE | ID: mdl-29983732

ABSTRACT

Cornus officinalis Sieb. et Zucc. is part of the genus Cornus of the family Cornaceae. Ripening and dry fruits (Corni Fructus) are recognized as an essential herb medicine in the traditional Chinese medicine (TCM) and have been widely used for over 2000 years. This review provides a comprehensive summary of Corni Fructus (CF), including the botany, phytochemistry, traditional use, and current pharmacological activities. According to the basic theory of TCM, CF usually participates in various Chinese medicinal formulae to exert the essential roles in replenishing liver and kidney, arresting seminal emission and sweat. Based on modern pharmacological studies, about 90 compounds have been isolated and identified from CF. In vivo and in vitro experimental studies indicate that CF exhibits extensive pharmacological activities including hypoglycemic, antioxidant, anti-inflammatory, anticancer, neuroprotective, hepatoprotective, and nephroprotective activities. However, only about 18% of chemical constituents in CF were tested. It means the potential pharmacological activities and clinical values of CF need to be further investigated.

7.
J Nat Med ; 72(4): 882-889, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29752578

ABSTRACT

Podolactones are a class of structural diverse diterpenoid lactones, mainly isolated from the Podocarpus species. Several bioactivities have been disclosed for podolactones, including cytotoxicity and anti-atherosclerosis. In this study, the seeds of P. nagi were isolated by comprehensive chromatographic methods to obtain three new podolatones, named nagilactone B 1-O-ß-D-glucoside (1), nagilactone N3 3-O-ß-D-glucoside (2), and 2-epinagilactone B (3), as well as a known compound, nagilactone B (4). Their structures were determined by analyses of NMR and HRESIMS data. Compounds 1 and 2 significantly inhibited nitric oxide (NO) production on LPS-stimulated RAW264.7 macrophages, with IC50 values of 0.18 ± 0.04 and 0.53 ± 0.03 µM, respectively. Indomethacin (IC50 4.21 ± 0.32 µM) was used as a positive control. Compound 1 suppressed the expression of inducible NO synthase (iNOS) in a concentration-dependent manner, mediating through inhibiting nuclear factor-κB (NF-κB) activity. This is the first report regarding the anti-inflammatory effect of podolactones, which could be potential anti-inflammatory agents.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Drugs, Chinese Herbal/chemistry , NF-kappa B/metabolism , Nitric Oxide/metabolism , Seeds/chemistry , Anti-Inflammatory Agents/pharmacology , Drugs, Chinese Herbal/pharmacology
8.
Eur J Pharmacol ; 830: 17-25, 2018 Jul 05.
Article in English | MEDLINE | ID: mdl-29680228

ABSTRACT

Non-small cell lung cancer (NSCLC) is one of the most common forms and leading causes of cancer-related mortality worldwide, and discovery of new effective drugs still remains imperative to improve the survival rate. Nagilactone E (NLE) is a natural product isolated from Podocarpus nagi seeds, which has been used as raw materials for edible oil and industrial oil extraction. This study aimed to investigate the anticancer potential of NLE against NSCLC A549 and NCI-H1975 cells. MTT assay revealed that NLE inhibited the proliferation of A549 and NCI-H1975 cells with IC50s of 5.18 ±â€¯0.49 and 3.57 ±â€¯0.29 µM, respectively. NLE treatment inhibited clone formation in both cancer cell lines. Cell cycle analysis indicated that NLE treatment effectively induced G2 phase cell cycle arrest in A549 and NCI-H1975 cells. NLE downregulated the phosphorylation of cdc2 (Tyr15) and cdc25C (Ser216) as well as the expression level of the protein kinase Wee1 in concentration- and time-dependent manners. In addition, NLE treatment decreased the protein level of Cyclin B1 as well as its nuclear localization, which might decrease the activity of the Cyclin B1/cdc2 complex and induce G2 phase arrest. Long-term NLE treatment also induced caspase-dependent cell apoptosis, as evidenced by increase in Annexin V positive cells and the cleavage of PARP. To sum, NLE inhibited proliferation, induced G2 phase arrest, and triggered caspase-dependent apoptosis in NSCLC cells, suggesting it to be a potential leading compound for cancer treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/metabolism , Cyclin B1/metabolism , Diterpenes/pharmacology , G2 Phase Cell Cycle Checkpoints/drug effects , Lung Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin B1/genetics , Down-Regulation , Humans
9.
J Nat Prod ; 80(7): 2110-2117, 2017 07 28.
Article in English | MEDLINE | ID: mdl-28719204

ABSTRACT

Nine new norditerpenoids and dinorditerpenoids, 2-oxonagilactone A (1), 7ß-hydroxynagilactone D (2), nagilactones K and L (3 and 4), 3ß-hydroxynagilactone L (5), 2ß-hydroxynagilactone L (6), 3-epi-15-hydroxynagilactone D (7), 1α-chloro-2ß,3ß,15-trihydroxynagilactone L (8), and 15-hydroxynagilactone L (9), were isolated from the seeds of Podocarpus nagi, along with eight known analogues. The structures of the new compounds were established based on detailed NMR and HRESIMS analysis, as well as from their ECD spectra. The absolute configuration of the known compound 1-deoxy-2α-hydroxynagilactone A (16) was confirmed by single-crystal X-ray diffraction. All of the isolates were tested for their cytotoxic activities against cancer cells. The results indicated that compounds 4 and 6, as well as several known compounds, displayed cytotoxicity against A2780 and HEY cancer cells. Among the new compounds, 2ß-hydroxynagilactone L (6) showed IC50 values of less than 2.5 µM against the two cell lines used. Furthermore, compound 6 induced autophagic flux in A2780 cells, as evidenced by an enhanced expression level of the autophagy marker phosphatidylethanolamine-modified microtubule-associated protein light-chain 3 (LC3-II) and increased mRFP-GFP-LC3 puncta. Also, compound 6 activated the c-Jun N-terminal kinase (JNK) pathway, while pretreatment with the JNK inhibitor SP600125 decreased compound 6-induced autophagy.


Subject(s)
Antineoplastic Agents, Phytogenic/isolation & purification , Antineoplastic Agents, Phytogenic/pharmacology , Diterpenes/isolation & purification , Diterpenes/pharmacology , Drugs, Chinese Herbal/isolation & purification , Drugs, Chinese Herbal/pharmacology , Seeds/chemistry , Anthracenes/chemistry , Antineoplastic Agents, Phytogenic/chemistry , Autophagy/drug effects , Cell Survival/drug effects , Cytotoxins , Diterpenes/chemistry , Drug Screening Assays, Antitumor , Drugs, Chinese Herbal/chemistry , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Molecular Structure , Nuclear Magnetic Resonance, Biomolecular
10.
Chin J Nat Med ; 15(12): 881-888, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29329644

ABSTRACT

Pharmaceutical research has focused on the discovery and development of anticancer drugs. Clinical application of chemotherapy drugs is limited due to their severe side effects. In this regard, new naturally occurring anticancer drugs have gained increasing attention because of their potential effectiveness and safety. Fruits and vegetables are promising sources of anticancer remedy. Clausena (family Rutaceae) is a genus of flowering plants and includes several kinds of edible fruits and vegetables. Phytochemical and pharmacological studies show that carbazole alkaloids and coumarins from Clausena plants exhibit anticancer activity. This review summarizes research progresses made in the anticancer properties of plants belonging to Clausena; in particular, compounds with direct cytotoxicity, cell cycle arrest, apoptosis induction, and immune potentiation effects are discussed. This review reveals the potential use of plants from Clausena in preventing and treating cancer and provides a basis for development of relevant therapeutic agents.


Subject(s)
Alkaloids/pharmacology , Antineoplastic Agents/pharmacology , Carbazoles/pharmacology , Clausena/chemistry , Coumarins/pharmacology , Plants, Medicinal/chemistry , Alkaloids/chemistry , Alkaloids/therapeutic use , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Carbazoles/chemistry , Carbazoles/therapeutic use , Cell Cycle Checkpoints/drug effects , Coumarins/chemistry , Coumarins/therapeutic use , Drugs, Chinese Herbal/chemistry , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/therapeutic use , Humans
11.
Planta Med ; 82(15): 1309-1328, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27542177

ABSTRACT

Diterpenoid dimers are rare in nature and mainly found in higher plants including the families Acanthaceae, Annonaceae, Asteraceae, Calceolariaceae, Chrysobalanaceae, Cupressaceae, Euphorbiaceae, Fabaceae, Lamiaceae, Liliaceae, Meliaceae, Rhizophoraceae, Taxaceae, Velloziaceae, and Zingiberaceae. In addition, a few diterpenoid dimers have been also reported from fungi (Psathyrellaceae), liverworts (Scapaniaceae), and a gorgonian (Gorgoniidae). They feature a wide variety of structures due to different core skeletons, linkage patterns, substituents, and configurations. Accordingly, diterpenoid dimers exhibit a broad range of bioactivities, including cytotoxic, anti-inflammatory, antimicrobial, antimalarial, and antifouling properties, which have attracted more and more research interests in the past decades. This review with 176 metabolites from 109 references provides a comprehensive and up-to-date overview of the source, biosynthesis, structure, synthesis, and bioactivities of diterpenoid dimers.


Subject(s)
Diterpenes/chemistry , Diterpenes/pharmacology , Anti-Infective Agents/chemistry , Anti-Infective Agents/pharmacology , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacology , Antimalarials/chemistry , Antimalarials/pharmacology , Diterpenes/chemical synthesis , Diterpenes/metabolism , Fabaceae/chemistry , Fungi , Liliaceae/chemistry , Meliaceae/chemistry , Molecular Structure , Rhizophoraceae/chemistry
12.
Acta Pharmacol Sin ; 28(11): 1783-90, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17959029

ABSTRACT

AIM: To investigate the role of death-associated protein kinase (DAPK) on the apoptosis of Raji cells induced by sodium butyrate. METHODS: The apoptosis of Raji cells were induced by sodium butyrate for 2, 4, 6, 8, and 10 d. Simultaneity, the Raji cells were inhibited to adhere on culture flask by polyHEME. Cell viability was detected by 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide method and the cell apoptosis percentage was estimated by flow cytometry. DAPK and focal adhesion kinase (FAK) expression were measured by Western blotting. Coding sequence on the C-terminal of DAPK, which can suppress the function of DAPK, was transfected into the Raji cells to investigate whether the C-terminal of DAPK could inhibit the apoptosis of Raji cells induced by sodium butyrate. RESULTS: After being treated with sodium butyrate, the Raji cells expressed DAPK and displayed many protrusions to adhere onto the culture flask. The Raji cells were susceptible to apoptosis when they were inhibited adhesion by polyHEME. At that time, the cell viability decreased, the cell apoptosis percentage increased and the protein levels of total FAK were reduced. The Raji cells, which were transfected with the coding region on the C-terminal of DAPK, sustained apoptosis and the FAK protein level when treated with sodium butyrate. CONCLUSION: Sodium butyrate induced DAPK expression. It caused the Raji cells to display many protrusions all around the cells and adhere onto the culture flask. DAPK expression prompted apoptosis by reducing the FAK protein level in sodium butyrate-induced Raji cells.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Burkitt Lymphoma/enzymology , Burkitt Lymphoma/ultrastructure , Butyrates/pharmacology , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Shape/drug effects , Focal Adhesion Kinase 1/metabolism , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Death-Associated Protein Kinases , Humans , In Vitro Techniques
13.
Ai Zheng ; 23(5): 497-501, 2004 May.
Article in Chinese | MEDLINE | ID: mdl-15142442

ABSTRACT

BACKGROUND & OBJECTIVE: Loss of activity of death- associated protein kinase 1 (DAPK1) may be an independent factor affecting survival of non-small cell lung cancer patients. DAPK1 over-expression can induce cell apoptosis and inhibit tumor cell metastasis. However, the mechanism of DAPK1 inhibiting metastasis was unclear yet. This study was designed to investigate the mechanism of DAPK1 affecting PGCl3 cells' growth and metastasis. METHODS: Open read frame (ORF) of DAPK1 gene was recombined into eukaryotic express vector pcDNA3.1. The PGCl3 cell line, a high metastasis lung tumor cell line, was transfected with pcDNA3.1-DAPK1 by lipofectamine2000. The growth curve of PGCl3 cells, colony formation assays, invasive, migration and adhesion ability were evaluated. Gelatinase secretion of PGCl3 cells treated with DAPK1 and expression of p53 and bcl-2 gene in PGCl3 cells were determined. RESULTS: PGCl3 cells of pcDNA3.1-DAPK1-transfected group grew slower than blank group and pcDNA3.1-transfected group. The numbers of colony formation of pcDNA3.1-DAPK1-transfected group reduced in comparison with blank group and pcDNA3.1-transfected group (P< 0.05). Invasive ability of pcDNA3.1-DAPK1-transfected group was 68.5% to blank group, and pcDNA3.1-transfected group was 88% to blank group. Migration ability of pcDNA3.1-DAPK1-transfected group was 87.3% to blank group and pcDNA3.1-transfected group was 95.7% to blank group. Adhesion ability of pcDNA3.1-DAPK1-transfected group was 62.7% to blank group, and pcDNA3.1-transfected group was 91.2% to blank group. Changes in PGCl3 cells secreting gelatinase have not been observed in different groups. Expression of p53 gene was upregulated in pcDNA3.1-DAPK1-transfected group, while expression of bcl-2 gene was downregulated. CONCLUSION: DAPK1 gene over-expression could suppress PGCl3 cells malignant phenotype, inhibit PGCl3 cells growth, invasive, migration and adhesion ability, upregulate p53 gene and downregulate bcl-2 gene. All of these changes may be the mechanism of inhibition of non-small cell lung cancer metastasis induced by DAPK1.


Subject(s)
Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/pathology , Protein Serine-Threonine Kinases , Transfection , Apoptosis Regulatory Proteins , Calcium-Calmodulin-Dependent Protein Kinases , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Adhesion , Cell Line, Tumor , Cell Movement , Cell Proliferation , Death-Associated Protein Kinases , Gene Expression Regulation, Neoplastic , Genes, bcl-2 , Genes, p53 , Genetic Vectors , Humans , Lung Neoplasms/metabolism , Neoplasm Invasiveness , Neoplasm Metastasis/physiopathology , Open Reading Frames , Protein Serine-Threonine Kinases/biosynthesis , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/physiology , Recombination, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...