Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 71
Filter
Add more filters

Complementary Medicines
Publication year range
1.
Genes (Basel) ; 14(9)2023 09 19.
Article in English | MEDLINE | ID: mdl-37761958

ABSTRACT

Previous studies have shown that inhibition of TNF family member FN14 (gene: TNFRSF12A) in colon tumors decreases inflammatory cytokine expression and mitigates cancer-induced cachexia. However, the molecular mechanisms underlying the regulation of FN14 expression remain unclear. Tumor microenvironments are often devoid of nutrients and oxygen, yet how the cachexic response relates to the tumor microenvironment and, importantly, nutrient stress is unknown. Here, we looked at the connections between metabolic stress and FN14 expression. We found that TNFRSF12A expression was transcriptionally induced during glutamine deprivation in cancer cell lines. We also show that the downstream glutaminolysis metabolite, alpha-ketoglutarate (aKG), is sufficient to rescue glutamine-deprivation-promoted TNFRSF12A induction. As aKG is a co-factor for histone de-methylase, we looked at histone methylation and found that histone H3K4me3 at the Tnfrsf12a promoter is increased under glutamine-deprived conditions and rescued via DM-aKG supplementation. Finally, expression of Tnfrsf12a and cachexia-induced weight loss can be inhibited in vivo by DM-aKG in a mouse cancer cachexia model. These findings highlight a connection between metabolic stress and cancer cachexia development.


Subject(s)
Cachexia , Colonic Neoplasms , TWEAK Receptor , Animals , Mice , Cachexia/genetics , Cachexia/prevention & control , Disease Models, Animal , Glutamine/pharmacology , Histone Code , Histone Methyltransferases , Histones/genetics , Ketoglutaric Acids/pharmacology , Tumor Microenvironment , Humans , Cell Line, Tumor/metabolism , TWEAK Receptor/genetics , TWEAK Receptor/metabolism
2.
Int J Mol Sci ; 22(7)2021 Mar 30.
Article in English | MEDLINE | ID: mdl-33808426

ABSTRACT

Dietary methyl-donors play important roles in physiological processes catalyzed by B vitamins as coenzymes, and are used for complementary support in oncotherapy. Our hypothesis was that methyl-donors can not only assist in tolerating cancer treatment but may also directly interfere with tumor growth and proliferation. Therefore, we investigated the proposed cancer inhibitory effects of methyl-donors (in a mixture of L-methionine, choline chloride, folic acid, and vitamin B12) on MCF7 and T47D breast cancer as well as A549 and H1650 lung cancer cell lines. Indeed, methyl-donor treatment significantly reduced the proliferation in all cell lines, possibly through the downregulation of MAPK/ERK and AKT signaling. These were accompanied by the upregulation of the pro-apoptotic Bak and Bax, both in MCF7 and H1650 cells, at reduced anti-apoptotic Mcl-1 and Bcl-2 levels in MCF7 and H1650 cells, respectively. The treatment-induced downregulation of p-p53(Thr55) was likely to contribute to protecting the nuclear localization and apoptosis inducing functions of p53. The presented features are known to improve the sensitivity of cancer therapy. Therefore, these data support the hypothesis, i.e., that methyl-donors may promote apoptotic signaling by protecting p53 functions through downregulating both the MAPK/ERK and the AKT pathways both in breast and lung adenocarcinoma cell lines. Our results can emphasize the importance and benefits of the appropriate dietary supports in cancer treatments. However, further studies are required to confirm these effects without any adverse outcome in clinical settings.


Subject(s)
Apoptosis/physiology , MAP Kinase Signaling System/physiology , Proto-Oncogene Proteins c-akt/metabolism , Apoptosis/drug effects , Breast/pathology , Breast Neoplasms/metabolism , Cell Line, Tumor/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Choline/pharmacology , Folic Acid/pharmacology , Humans , Lung/pathology , Lung Neoplasms/metabolism , MAP Kinase Signaling System/drug effects , Methionine/pharmacology , Methylation/drug effects , Proto-Oncogene Proteins c-akt/drug effects , Signal Transduction/drug effects , Vitamin B 12/pharmacology
3.
Theranostics ; 10(5): 2260-2272, 2020.
Article in English | MEDLINE | ID: mdl-32104506

ABSTRACT

Semiconducting polymers (SPs)-based dual photothermal therapy (PTT) obtained better therapeutic effect than single PTT due to its higher photothermal conversion efficiency. However, most dual PTT need to use two lasers for heat generation, which brings about inconvenience and limitation to the experimental operations. Herein, we report the development of "nanococktail" nanomaterials (DTPR) with 808 nm-activated image-guided dual photothermal properties for optimized cancer therapy. Methods: In this work, we co-encapsulated AIEgens (TPA-BDTO, T) and SPs (PDPPP, P) by using maleimide terminated amphiphilic polymer (DSPE-PEG2000-Mal, D), then further conjugated the targeting ligands (RGD, R) through "click" reaction. Finally, such dual PTT nanococktail (termed as DTPR) was constructed. Results: Once DTPR upon irradiation with 808 nm laser, near-infrared fluorescence from T could be partially converted into thermal energy through fluorescence resonance energy transfer (FRET) between T and P, coupling with the original heat energy generated by the photothermal agent P itself, thus resulting in image-guided dual PTT. The photothermal conversion efficiency of DTPR reached 60.3% (dual PTT), much higher as compared to its inherent photothermal effect of only 31.5% (single PTT), which was further proved by the more severe photothermal ablation in vitro and in vivo upon 808 nm laser irradiation. Conclusion: Such smart "nanococktail" nanomaterials could be recognized as a promising photothermal nanotheranostics for image-guided cancer treatment.


Subject(s)
Fluorescence Resonance Energy Transfer/instrumentation , Photothermal Therapy/methods , Theranostic Nanomedicine/methods , Animals , Cell Line, Tumor/drug effects , Cell Line, Tumor/metabolism , Cell Line, Tumor/radiation effects , Drug Delivery Systems/methods , Fluorescence , Hyperthermia, Induced/methods , Lasers , Ligands , Mice , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Polymers , Semiconductors
4.
Theranostics ; 10(4): 1555-1571, 2020.
Article in English | MEDLINE | ID: mdl-32042322

ABSTRACT

As a hallmark of metabolic reprogramming, aerobic glycolysis contributes to tumorigenesis and aggressiveness. However, the mechanisms and therapeutic strategies regulating aerobic glycolysis in neuroblastoma (NB), one of leading causes of cancer-related death in childhood, still remain elusive. Methods: Transcriptional regulators and their downstream glycolytic genes were identified by a comprehensive screening of publicly available datasets. Dual-luciferase, chromatin immunoprecipitation, real-time quantitative RT-PCR, western blot, gene over-expression or silencing, co-immunoprecipitation, mass spectrometry, peptide pull-down assay, sucrose gradient sedimentation, seahorse extracellular flux, MTT colorimetric, soft agar, matrigel invasion, and nude mice assays were undertaken to explore the biological effects and underlying mechanisms of transcriptional regulators in NB cells. Survival analysis was performed by using log-rank test and Cox regression assay. Results: Transcription factor myeloid zinc finger 1 (MZF1) was identified as an independent prognostic factor (hazard ratio=2.330, 95% confidence interval=1.021 to 3.317), and facilitated glycolysis process through increasing expression of hexokinase 2 (HK2) and phosphoglycerate kinase 1 (PGK1). Meanwhile, a 21-amino acid peptide encoded by upstream open reading frame of MZF1, termed as MZF1-uPEP, bound to zinc finger domain of Yin Yang 1 (YY1), resulting in repressed transactivation of YY1 and decreased transcription of MZF1 and downstream genes HK2 and PGK1. Administration of a cell-penetrating MZF1-uPEP or lentivirus over-expressing MZF1-uPEP inhibited the aerobic glycolysis, tumorigenesis and aggressiveness of NB cells. In clinical NB cases, low expression of MZF1-uPEP or high expression of MZF1, YY1, HK2, or PGK1 was associated with poor survival of patients. Conclusions: These results indicate that therapeutic targeting of YY1/MZF1 axis by MZF1-uPEP inhibits aerobic glycolysis and NB progression.


Subject(s)
Molecular Targeted Therapy/methods , Neuroblastoma/drug therapy , Warburg Effect, Oncologic/drug effects , Animals , Cell Line, Tumor/drug effects , Cell Line, Tumor/metabolism , Cell Proliferation/genetics , Child , Disease Progression , Female , Gene Expression Regulation, Neoplastic , Hexokinase/metabolism , Humans , Kruppel-Like Transcription Factors/drug effects , Kruppel-Like Transcription Factors/metabolism , Mice , Mice, Nude , Neuroblastoma/metabolism , Neuroblastoma/pathology , Promoter Regions, Genetic , Survival Analysis , Transcription Factors/drug effects , Transcription Factors/metabolism , YY1 Transcription Factor/drug effects , YY1 Transcription Factor/metabolism
5.
Sci Rep ; 9(1): 16647, 2019 11 12.
Article in English | MEDLINE | ID: mdl-31719636

ABSTRACT

The present state of cancer chemotherapy is unsatisfactory. New anticancer drugs that marginally improve the survival of patients continue to be developed at an unsustainably high cost. The study aimed to elucidate the effects of insulin (INS), an inexpensive drug with a convincing safety profile, on the susceptibility of colon cancer to chemotherapeutic agents: 5-fluorouracil (FU), oxaliplatin (OXA), irinotecan (IRI), cyclophosphamide (CPA) and docetaxel (DOC). To examine the effects of insulin on cell viability and apoptosis, we performed an in vitro analysis on colon cancer cell lines Caco-2 and SW480. To verify the results, we performed in vivo analysis on mice bearing MC38 colon tumors. To assess the underlying mechanism of the therapy, we examined the mRNA expression of pathways related to the signaling downstream of insulin receptors (INSR). Moreover, we performed Western blotting to confirm expression patterns derived from the genetic analysis. For the quantification of circulating tumor cells in the peripheral blood, we used the maintrac method. The results of our study show that insulin-pretreated colon cancer cells are significantly more susceptible to commonly used chemotherapeutics. The apoptosis ratio was also enhanced when INS was administered complementary to the examined drugs. The in vivo study showed that the combination of INS and FU resulted in significant inhibition of tumor growth and reduction of the number of circulating tumor cells. This combination caused a significant downregulation of the key signaling substrates downstream of INSR. The results indicate that the downregulation of PIK3CA (phosphatidylinositol 3-kinase catalytic subunit alpha), which plays a critical role in cell signaling and GRB2 (growth factor receptor-bound protein 2), a regulator of cell proliferation and differentiation may be responsible for the sensitizing effect of INS. These findings were confirmed at protein levels by Western blotting. In conclusion, these results suggest that INS might be potentially applied to clinical use to enhance the therapeutic effectiveness of chemotherapeutic drugs. The findings may become a platform for the future development of new and inexpensive strategies for the clinical chemotherapy of tumors.


Subject(s)
Antineoplastic Agents/therapeutic use , Class I Phosphatidylinositol 3-Kinases/antagonists & inhibitors , Colorectal Neoplasms/drug therapy , GRB2 Adaptor Protein/antagonists & inhibitors , Insulin/pharmacology , Animals , Blotting, Western , Caco-2 Cells/drug effects , Caco-2 Cells/metabolism , Cell Line, Tumor/drug effects , Cell Line, Tumor/metabolism , Class I Phosphatidylinositol 3-Kinases/metabolism , Colorectal Neoplasms/metabolism , Cyclophosphamide/therapeutic use , Docetaxel/therapeutic use , Down-Regulation/drug effects , Drug Synergism , Female , Fluorouracil/therapeutic use , GRB2 Adaptor Protein/metabolism , Humans , Irinotecan/therapeutic use , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Neoplastic Cells, Circulating/drug effects , Neoplastic Cells, Circulating/metabolism , Oxaliplatin/therapeutic use
6.
Medicina (Kaunas) ; 54(2)2018 Mar 30.
Article in English | MEDLINE | ID: mdl-30344242

ABSTRACT

Background and objectives: Cell culture is one of the mainstays in the research of breast cancer biology, although the extent to which this approach allows to preserve the original characteristics of originating tumor and implications of cell culture findings to real life situations have been widely debated in the literature. The aim of this study was to determine the role of three cell culture media on transcriptional expression of breast cancer markers in three breast cancer reference cell lines (MCF7, SkBr3 and MDA-MB-436). Materials and methods: Cell lines were conditioned in three studied media (all containing 5% fetal bovine serum (FBS) + hormones/growth factors; different composition of basal media) for four passages. Population growth was characterized by cumulative population doubling levels, average generation time, cell yield and viability at the fourth passage. Transcriptional expression of breast cancer differentiation markers and regulatory transcriptional programs was measured by qPCR. Results: Differences in the composition of growth media significantly influenced the growth of studied cell lines and the expression of mammary lineage governing transcriptional programs and luminal/basal markers. Effects of media on transcriptional expression were more pronounced in luminal cell lines (MCF7, SkBr3), than in the basal cell line (MDA-MB-436). Changes in growth media in terms of supplementation and basal medium delayed growth of cells, but improved cell yields. Conclusions: The expression of breast cancer cell differentiation phenotypic markers depends on the composition of cell growth medium, therefore cell culture as a tool in phenotypic studies should be used considering this effect. The findings of such studies should always be interpreted with caution. The formulation of cell growth media has greater effect on the expression of phenotypic markers in luminal, rather than basal cell lines. Media containing mitogens and higher vitamin content improved efficacy of cell culture in terms of cell yields, although greatly increased growth times.


Subject(s)
Biomarkers, Tumor/genetics , Breast Neoplasms/genetics , Cell Differentiation/drug effects , Cell Line, Tumor/drug effects , Culture Media, Conditioned/pharmacology , Gene Expression Profiling/methods , Biomarkers, Tumor/metabolism , Breast Neoplasms/pathology , Cell Differentiation/genetics , Cell Line, Tumor/metabolism , Cell Line, Tumor/pathology , Culture Media, Conditioned/chemistry , Female , Humans , MCF-7 Cells/drug effects , MCF-7 Cells/metabolism , MCF-7 Cells/pathology , Reverse Transcriptase Polymerase Chain Reaction
7.
J Med Food ; 21(12): 1244-1252, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30153075

ABSTRACT

Sweet olive (Osmanthus fragrans flowers) is used to treat dysentery and reduce phlegm and stasis in traditional Chinese medicine. Recently, we found that verbascoside, the major component in the sweet olive ethanolic extract (OFE), inhibited IL-8 secretion in human colorectal adenocarcinoma WiDr cells. However, evidence-based treatment of inflammatory bowel disease (IBD) with the extract is yet to be performed. To evaluate the therapeutic effect of OFE, we measured IL-8 suppression by OFE and verbascoside in a WiDr cell culture assay. In the IL-8 secretion assay, both OFE (100 µg/mL) and verbascoside (10 µM) significantly inhibited IL-8 production in WiDr cells. Furthermore, we designed cotreated (dextran sulfate sodium [DSS]+OFE-treated) and post-treated (DSS-OFE-treated) protocols to access the therapeutic effects of OFE in vivo. Mice treated with 500 mg/kg per day OFE exhibited significant improvement in IBD symptoms, including disease activity index score, body weight, and colon length maintenance. The suppressive effects on myeloperoxidase expression and lower histopathology scores (including neutrophil infiltration) for the colon were also found. These findings suggest that OFE exerts anti-inflammatory effect on DSS-induced colitis.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Biological Products/therapeutic use , Colitis/drug therapy , Olea , Phytotherapy , Animals , Anti-Inflammatory Agents/pharmacology , Biological Products/pharmacology , Cell Line, Tumor/drug effects , Cell Line, Tumor/metabolism , Colitis/chemically induced , Dextran Sulfate , Disease Models, Animal , Humans , Interleukin-8/metabolism , Mice , Peroxidase/metabolism
8.
Haematologica ; 103(11): 1760-1771, 2018 11.
Article in English | MEDLINE | ID: mdl-29976735

ABSTRACT

Mastocytosis is a term used to denote a group of rare diseases characterized by an abnormal accumulation of neoplastic mast cells in various tissues and organs. In most patients with systemic mastocytosis, the neoplastic cells carry activating mutations in KIT Progress in mastocytosis research has long been hindered by the lack of suitable in vitro models, such as permanent human mast cell lines. In fact, only a few human mast cell lines are available to date: HMC-1, LAD1/2, LUVA, ROSA and MCPV-1. The HMC-1 and LAD1/2 cell lines were derived from patients with mast cell leukemia. By contrast, the more recently established LUVA, ROSA and MCPV-1 cell lines were derived from CD34+ cells of non-mastocytosis donors. While some of these cell lines (LAD1/2, LUVA, ROSAKIT WT and MCPV-1) do not harbor KIT mutations, HMC-1 and ROSAKIT D816V cells exhibit activating KIT mutations found in mastocytosis and have thus been used to study disease pathogenesis. In addition, these cell lines are increasingly employed to validate new therapeutic targets and to screen for effects of new targeted drugs. Recently, the ROSAKIT D816V subclone has been successfully used to generate a unique in vivo model of advanced mastocytosis by injection into immunocompromised mice. Such a model may allow in vivo validation of data obtained in vitro with targeted drugs directed against mastocytosis. In this review, we discuss the major characteristics of all available human mast cell lines, with particular emphasis on the use of HMC-1 and ROSAKIT D816V cells in preclinical therapeutic research in mastocytosis.


Subject(s)
Cell Line, Tumor , Mast Cells , Mastocytosis, Systemic , Models, Biological , Animals , Cell Line, Tumor/metabolism , Cell Line, Tumor/pathology , Humans , Mast Cells/metabolism , Mast Cells/pathology , Mastocytosis, Systemic/genetics , Mastocytosis, Systemic/metabolism , Mastocytosis, Systemic/pathology
9.
Res Vet Sci ; 114: 461-468, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28802138

ABSTRACT

Feline mammary carcinoma (FMC) is a highly aggressive pathology that has been proposed as an interesting model of breast cancer disease, especially for the hormone refractory subgroup. Recently, cancer cell metabolism has been described as a hallmark of cancer cells. Here, we investigate the effects and mechanism of metabolic modulation by metformin (MET, anti-diabetic drug), 2-deoxyglucose (2DG, hexokinase inhibitor) or a combination of both drugs, MET/2DG on two established FMC cells lines: AlRB (HER2 (3+) and Ki67<5%) and AlRATN (HER2 (-) and Ki67>15%). We found that treatments significantly decreased both FMC cells viability by up to 80%. AlRB resulted more sensitive to 2DG than AlRATN (IC50: 3.15 vs 6.32mM, respectively). The combination of MET/2DG potentiated the effects of the individually added drugs on FMC cells. In addition, MET/2DG caused an increased in intracellular oxidants, autophagic vesicles and completely inhibited colony formation. Conversely, only MET significantly altered plasma membrane integrity, presented late apoptotic/necrotic cells and increased both glucose consumption and lactate concentration. Our results support further studies to investigate the potential use of this metabolic modulation approach in a clinical veterinary setting.


Subject(s)
Antineoplastic Agents/pharmacology , Deoxyglucose/metabolism , Energy Metabolism/drug effects , Mammary Neoplasms, Animal/metabolism , Metformin/pharmacology , Animals , Cats , Cell Line, Tumor/metabolism , Cell Survival/drug effects
10.
PLoS One ; 10(9): e0136542, 2015.
Article in English | MEDLINE | ID: mdl-26325577

ABSTRACT

Omega 3 (n3) and Omega 6 (n6) polyunsaturated fatty acids (PUFAs) have been reported to exhibit opposing roles in cancer progression. Our objective was to determine whether different ratios of n6/n3 (AA/EPA+DHA) FAs could modulate the cell viability, lipid peroxidation, total cellular fatty acid composition and expression of tumor regulatory Matrix Attachment Region binding proteins (MARBPs) in breast cancer cell lines and in non-cancerous, MCF10A cells. Low ratios of n6/n3 (1:2.5, 1:4, 1:5, 1:10) FA decreased the viability and growth of MDA-MB-231 and MCF7 significantly compared to the non-cancerous cells (MCF10A). Contrarily, higher n6/n3 FA (2.5:1, 4:1, 5:1, 10:1) decreased the survival of both the cancerous and non-cancerous cell types. Lower ratios of n6/n3 selectively induced LPO in the breast cancer cells whereas the higher ratios induced in both cancerous and non-cancerous cell types. Interestingly, compared to higher n6/n3 FA ratios, lower ratios increased the expression of tumor suppressor MARBP, SMAR1 and decreased the expression of tumor activator Cux/CDP in both breast cancer and non-cancerous, MCF10A cells. Low n6/n3 FAs significantly increased SMAR1 expression which resulted into activation of p21WAF1/CIP1 in MDA-MB-231 and MCF7, the increase being ratio dependent in MDA-MB-231. These results suggest that increased intake of n3 fatty acids in our diet could help both in the prevention as well as management of breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Fatty Acids, Omega-3/pharmacology , Fatty Acids, Omega-6/pharmacology , Lipid Peroxidation , MCF-7 Cells/metabolism , Matrix Attachment Region Binding Proteins/metabolism , Blotting, Western , Breast Neoplasms/chemistry , Cell Line, Tumor/chemistry , Cell Line, Tumor/drug effects , Cell Line, Tumor/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Docosahexaenoic Acids/pharmacology , Dose-Response Relationship, Drug , Eicosapentaenoic Acid/pharmacology , Fatty Acids, Omega-3/analysis , Fatty Acids, Omega-6/analysis , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , MCF-7 Cells/chemistry , MCF-7 Cells/drug effects , alpha-Linolenic Acid/pharmacology
11.
Urology ; 82(6): 1197-203, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23992971

ABSTRACT

The US diet has been fortified with folic acid to prevent neural tube defects since 1998. The Physician Data Queries from the National Cancer Institute describe folate as protective against prostate cancer, whereas its synthetic analog, folic acid, is considered to increase prostate cancer risk when taken at levels easily achievable by eating fortified food or taking over-the-counter supplements. We review the present literature to examine the effects of folate and folic acid on prostate cancer, help interpret previous epidemiologic data, and provide clarification regarding the apparently opposing roles of folate for patients with prostate cancer. A literature search was conducted in Medline to identify studies investigating the effect of nutrition and specifically folate and folic acid on prostate carcinogenesis and progression. In addition, the National Health and Nutrition Examination Survey database was analyzed for trends in serum folate levels before and after mandatory fortification. Folate likely plays a dual role in prostate carcinogenesis. There remains conflicting epidemiologic evidence regarding folate and prostate cancer risk; however, there is growing experimental evidence that higher circulating folate levels can contribute to prostate cancer progression. Further research is needed to clarify these complex relationships.


Subject(s)
Prostatic Neoplasms/physiopathology , Animals , Carcinogenesis/genetics , Carcinogenesis/metabolism , Cell Line, Tumor/metabolism , Disease Progression , Folic Acid/blood , Folic Acid/physiology , Folic Acid Deficiency/epidemiology , Humans , Immunohistochemistry , Kallikreins/metabolism , Kallikreins/physiology , Male , Nutrition Surveys , Prostate-Specific Antigen/metabolism , Prostate-Specific Antigen/physiology , Prostatic Neoplasms/epidemiology , Prostatic Neoplasms/metabolism , Vitamin B Complex/physiology
12.
Environ Toxicol Pharmacol ; 35(2): 325-34, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23434523

ABSTRACT

The medicinal plants Aristolochia clematitis L. as well as Asarum europaeum L., representatives of the plant family Aristolochiaceae and mentioned in the German Homeopathic Pharmacopeia, contain aristolochic acid. We found that the mother tinctures of A. clematitis and A. europaeum inhibited DNA synthesis in human hepatoma HepG2 cells in a dose-dependent manner. One of the components of the plant extract, aristolochic acid I (AAI), is linked to the development of nephropathy and urothelial cancer in humans. Therefore, we also evaluated the cytotoxicity and genotoxicity of AAI in HepG2 cells. Cell proliferation was inhibited concentration-dependently by AAI using BrdU-ELISA and colony forming assay. AAI formed DNA adducts (measured by (32)P-postlabeling), induced chromosomal aberrations (micronuclei) and DNA strand breaks. DNA damage induced by AAI led to an arrest of cells in the S-phase which was associated with the increased expression of p53 and p21 proteins. The results are discussed under consideration of former studies.


Subject(s)
Aristolochiaceae/chemistry , Aristolochic Acids/toxicity , Cell Line, Tumor/drug effects , Cell Line, Tumor/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , DNA Adducts/analysis , DNA Damage/drug effects , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Homeopathy/methods , Humans , Mutagenicity Tests/methods , NAD(P)H Dehydrogenase (Quinone)/metabolism , Plant Extracts/chemistry , Plant Extracts/toxicity , S Phase Cell Cycle Checkpoints/drug effects
13.
Zhong Xi Yi Jie He Xue Bao ; 8(10): 968-73, 2010 Oct.
Article in Chinese | MEDLINE | ID: mdl-20939988

ABSTRACT

OBJECTIVE: To investigate the effects of Jianpi Jiedu Formula (JPJDF), a compound Chinese herbal medicine, on vascular endothelial growth factor (VEGF) expression induced by Helicobacter pylori (Hp) in human gastric cancer cells, and to explore the possible mechanism. METHODS: Human gastric MKN45 cancer cells were infected with standard Hp NCTC11637 by coculture, and the cells were divided into 7 groups: normal control group, Hp group, NS398 inhibitor group, blank serum group, and 5%, 10% and 20% JPJDF groups. The expressions of VEGF and cyclooxygenase-2 (COX-2) mRNAs in human gastric cancer cells infected by Hp were evaluated by real-time fluorogenic quantitative polymerase chain reaction (PCR). After inhibiting the expression of COX-2 with COX-2 specific inhibitor NS398 (50 µmol/L), VEGF mRNA expression induced by Hp in human gastric cancer MKN45 cells was evaluated by real-time fluorogenic quantitative PCR. RESULTS: Real-time fluorogenic quantitative PCR results showed that COX-2 mRNA expression increased significantly after MKN45 cells were infected with Hp for 6 h (P<0.01), and VEGF mRNA expression also increased significantly after MKN45 cells were infected with Hp for 12 h as compared with the normal control group (P<0.01). After inhibiting COX-2 expression with COX-2 specific inhibitor NS398, Hp-induced VEGF mRNA expression significantly reduced (P<0.01). 5%, 10% and 20% JPJDF-containing sera all down-regulated VEGF and COX-2 mRNA expressions induced by Hp in a dose-dependent manner as compared with the Hp infection group. CONCLUSION: Hp could induce the expressions of COX-2 and VEGF in human gastric cancer cells, and JPJDF down-regulates Hp-induced expression of VEGF by inhibiting COX-2, which might be one of the important mechanisms of its prevention and treatment of gastric cancer.


Subject(s)
Cyclooxygenase 2 Inhibitors/pharmacology , Cyclooxygenase 2/metabolism , Drugs, Chinese Herbal/pharmacology , Helicobacter Infections/metabolism , Vascular Endothelial Growth Factor A/metabolism , Cell Line, Tumor/metabolism , Cell Line, Tumor/microbiology , Helicobacter pylori/pathogenicity , Humans
14.
World J Gastroenterol ; 16(27): 3377-84, 2010 Jul 21.
Article in English | MEDLINE | ID: mdl-20632439

ABSTRACT

AIM: To investigate the mechanism by which galangin, a polyphenolic compound derived from medicinal herbs, induces apoptosis of hepatocellular carcinoma (HCC) cells. METHODS: The 3-(4,5-dimethyl-thiazol-2-yl)-2,5-diphenyl-tetrazolium bromide assay was used to measure cell viability. Apoptosis was evaluated by in situ uptake of propidium iodide and Hoechst 33258 and was then detected by fluorescence microscopy. Protein expressions were detected by Western blotting. To confirm the apoptotic pathway mediated by galangin, cells were transfected by bcl-2 gene to overexpress Bcl-2 or siRNA to down-regulate Bcl-2 expression. RESULTS: Galangin (46.25-370.0 micromol/L) exerted an anti-proliferative effect, induced apoptosis, and decreased mitochondrial membrane potential in a dose and time-dependent manner. Treatment with galangin induced apoptosis by translocating the pro-apoptotic protein Bax to the mitochondria, which released apoptosis-inducing factor and cytochrome c into the cytosol. Overexpression of Bcl-2 attenuated galangin-induced HepG2 cell apoptosis, while decreasing Bcl-2 expression enhanced galangin-induced cell apoptosis. CONCLUSION: Our data suggests that galangin mediates apoptosis through a mitochondrial pathway, and may be a potential chemotherapeutic drug for the treatment of HCC.


Subject(s)
Apoptosis/drug effects , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor/drug effects , Flavonoids/pharmacology , Liver Neoplasms/pathology , Mitochondria/drug effects , Mutagens/pharmacology , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor/metabolism , Cell Line, Tumor/pathology , Dose-Response Relationship, Drug , Flavonoids/therapeutic use , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Membrane Potential, Mitochondrial/drug effects , Mitochondria/metabolism , Mutagens/therapeutic use , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology
15.
Genomics ; 94(6): 407-13, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19720140

ABSTRACT

Charcot-Marie-Tooth disease (CMT) is a heritable neurodegenerative condition, some types of which (notably CMT4A) are caused by mutations in the GDAP1 gene that encodes a protein of unknown molecular function implicated in regulation of mitochondrial fission. Here we present for the first time a functional analysis of the GDAP1 gene promoter which we found to be transcriptionally regulated by YY1, a broadly studied factor that seems to be involved in regulating many of the same cellular phenomena as GDAP1. We show that GDAP1 is broadly expressed in cancer cell lines of different tissue origin, contrasting with the restricted neuronal distribution reported by some authors. There is a consensus YY1 binding site in the GDAP1 core promoter which we show to be functional in both in vitro binding assays and in living cells. Overexpression of YY1 activated the GDAP1 promoter in a reporter gene system as well as increased the level of endogenous mRNA. RNAi-mediated knockdown of YY1 in HEK293 cells led to decreased GDAP1 expression. While YY1 is known to exert both positive and negative regulatory influences on nuclear-encoded mitochondrial proteins, as well as on neurodegeneration-related genes, in all cell lines we studied (including neuroblastoma) the effect of YY1 on GDAP1 expression is activatory. This leads to interesting conclusions about the possible clinical role of this interaction and suggests a broader regulatory network.


Subject(s)
Gene Expression Regulation/genetics , Nerve Tissue Proteins/genetics , Transcription, Genetic/genetics , YY1 Transcription Factor/physiology , Animals , Binding Sites , Cell Line, Tumor/metabolism , Computational Biology , Female , Gene Knockdown Techniques , Genes, Reporter , Humans , Mammals/genetics , Mitochondria/physiology , Mutagenesis, Site-Directed , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Nerve Tissue Proteins/biosynthesis , Organ Specificity , Promoter Regions, Genetic/genetics , RNA, Messenger/biosynthesis , RNA, Neoplasm/biosynthesis , Sequence Alignment , Sequence Homology, Nucleic Acid , Up-Regulation
16.
Cancer Biol Ther ; 8(14): 1389-401, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19483472

ABSTRACT

Despite recent advances in cancer therapies, metastatic renal cell carcinoma (RCC) remains difficult to treat. Most RCCs result from inactivation of the von Hippel Lindau (VHL) tumor suppressor, leading to stable expression of Hypoxia-Inducible Factor-alpha (HIF-1alpha, -2alpha, -3alpha) and the induction of downstream target genes, including those responsible for angiogenesis and metastasis. While VHL is inactivated in the majority of RCC cases, expression of the PTEN tumor suppressor is reduced in about 30% of cases. PTEN functions to antagonize PI3K/Akt/mTOR signaling, thereby controlling cell growth and survival. Activation of PI3K/Akt/mTOR leads to increased HIF-1alpha expression in certain cancer cells, supporting the rationale of using mTOR inhibitors as anti-cancer agents. Notably, HIF-2alpha, rather than HIF-1alpha, has been shown to play a critical role in renal tumorigenesis. To investigate whether HIF-2alpha is similarly regulated by the PI3K pathway in VHL(-/-)RCC cells, we manipulated PI3K signaling using PTEN overexpression and siRNA knockdown studies and pharmacologic inhibition of PI3K or Akt. Our data support a novel role for wild-type PTEN in promoting HIF-2alpha activity in VHL null RCC cells. This mechanism is unique to the cellular environment in which HIF-2alpha expression is deregulated, resulting from the loss of VHL function. Our data show that PTEN induces HIF-2alpha transcriptional activity by inhibiting expression of Yin Yang 1 (YY1), which acts as a novel corepressor of HIF-2alpha. Further, PTEN suppression of YY1 is mediated through antagonism of PI3K signaling. We conclude that wild-type PTEN relieves the repressive nature of YY1 at certain HIF-2alpha target promoters and that this mechanism may promote early renal tumorigenesis resulting from VHL inactivation by increasing HIF-2alpha activity.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Carcinoma, Renal Cell/pathology , Kidney Neoplasms/pathology , Neoplasm Proteins/physiology , PTEN Phosphohydrolase/physiology , Von Hippel-Lindau Tumor Suppressor Protein/physiology , YY1 Transcription Factor/antagonists & inhibitors , Basic Helix-Loop-Helix Transcription Factors/genetics , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Cell Line, Tumor/metabolism , Cell Transformation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic , Humans , Intracellular Signaling Peptides and Proteins/physiology , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Matrix Metalloproteinase 14/biosynthesis , Matrix Metalloproteinase 14/genetics , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Phosphatidylinositol 3-Kinases/physiology , Protein Serine-Threonine Kinases/physiology , Proto-Oncogene Proteins c-akt/physiology , Signal Transduction/genetics , TOR Serine-Threonine Kinases , Von Hippel-Lindau Tumor Suppressor Protein/genetics , YY1 Transcription Factor/physiology
17.
World J Gastroenterol ; 15(15): 1816-20, 2009 Apr 21.
Article in English | MEDLINE | ID: mdl-19370777

ABSTRACT

AIM: To investigate the inhibitory effect of acetylshikonin on human gastric carcinoma cell line SGC-7901 and its mechanism. METHODS: MTT assay was used to assess the inhibitory effect of acetylshikonin on proliferation of SGC-7901 cells. Apoptosis-inducing effect was determined by flow cytometry and terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end-labeling with Hoechst staining. Expression of mRNA and protein in Bcl-2 and Bax was analyzed by reverse transcription-polymerase chain reaction and Western blot. Antitumor effect of acetylshikonin on a mouse SGC-7901 model was also determined. RESULTS: Forty-eight hours after treatment with acetylshikonin, MTT assay showed that acetylshikonin inhibited the proliferation of SGC-7901 cells in a dose-dependent manner. The half maximal inhibitory concentration of acetylshikonin to SGC-7901 cells was 0.428 +/- 0.07 mg/L. Cell shrinkage, nuclear pyknosis and chromatin condensation, which are the characteristics of cell apoptosis, were observed in treated SGC-7901 cells and the percentage of apoptosis increased in a dose-dependent manner. Acetylshikonin down-regulated the expression of Bcl-2 and up-regulated the expression of Bax in the treated SGC-7901 cells compared with the controls. The experiment in vivo showed that 0.5, 1, and 2 mg/kg of acetylshikonin significantly inhibited the growth of tumor in the mouse SGC-7901 model, with an inhibitory rate of 25.00%-55.76%. CONCLUSION: Acetylshikonin inhibits the growth of SGC-7901 cells in vitro and in vivo by inducing cell apoptosis.


Subject(s)
Anthraquinones/pharmacology , Cell Line, Tumor/drug effects , Drugs, Chinese Herbal/pharmacology , Stomach Neoplasms , Animals , Caspase 3/metabolism , Cell Cycle/drug effects , Cell Line, Tumor/metabolism , Cell Proliferation/drug effects , Humans , Male , Mice , Mice, Nude , Neoplasm Transplantation , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
18.
Cancer Lett ; 281(1): 108-16, 2009 Aug 18.
Article in English | MEDLINE | ID: mdl-19332363

ABSTRACT

We examined the anti-angiogenic effects of Ganoderma tsugae methanol extract (GTME) on human epidermoid carcinoma A-431 cells. Our data indicate that GTME inhibits the expression of epidermal growth factor receptor (EGFR) and vascular endothelial growth factor (VEGF) in vitro and in vivo, and also inhibits the capillary tube formation of human umbilical vein endothelial cells (HUVECs). We also show that the suppression of VEGF expression by GTME can be restored by treatment with EGF. These results suggest that GTME inhibits VEGF expression via the suppression of EGFR expression, resulting in the downregulation of VEGF secretion from epidermoid carcinoma A-431 cells. These findings reveal a novel role for G. tsugae in inhibiting EGFR and VEGF expression, which are important for tumor angiogenesis and growth. Thus, GTME may provide a potential therapeutic approach for anti-tumor treatment.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Carcinoma, Squamous Cell/pathology , Drugs, Chinese Herbal/pharmacology , ErbB Receptors/biosynthesis , Ganoderma/chemistry , Neoplasm Proteins/biosynthesis , Neovascularization, Pathologic/drug therapy , Phytotherapy , Plant Extracts/pharmacology , Skin Neoplasms/pathology , Vascular Endothelial Growth Factor A/biosynthesis , Angiogenesis Inhibitors/therapeutic use , Animals , Antineoplastic Agents, Phytogenic/therapeutic use , Carcinoma, Squamous Cell/blood supply , Carcinoma, Squamous Cell/drug therapy , Cell Line, Tumor/drug effects , Cell Line, Tumor/metabolism , Cell Line, Tumor/transplantation , Cells, Cultured/drug effects , Drug Resistance, Neoplasm/drug effects , Drugs, Chinese Herbal/therapeutic use , Endothelium, Vascular/cytology , Epidermal Growth Factor/pharmacology , ErbB Receptors/genetics , ErbB Receptors/physiology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Methanol , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neovascularization, Pathologic/physiopathology , Neovascularization, Physiologic/drug effects , Plant Extracts/therapeutic use , Reishi , Skin Neoplasms/blood supply , Skin Neoplasms/drug therapy , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Xenograft Model Antitumor Assays
19.
Mol Cancer Ther ; 8(3): 655-64, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19240161

ABSTRACT

As cellular folate levels seem to have a different effect on cancer cells from different origins, we extended our initial study to a broader panel of cancer cells. BCRP and MRP1-5 expression was determined in KB, OVCAR-3, IGROV-1, ZR75-1/R/MTX, SCC-11B, SCC-22B, and WiDr either grown in standard RPMI 1640 containing 2.3 micromol/L supraphysiologic concentration of folic acid [high folate (HF)] or adapted to more physiologic concentrations [1-5 nmol/L folic acid or leucovorin; low folate (LF)]. Compared with the HF counterparts, KB LF cells displayed 16.1-fold increased MRP3 and OVCAR-3 LF cells showed 4.8-fold increased MRP4 mRNA levels along with increased MRP3 and MRP4 protein expression, respectively. A marked increase on BCRP protein and mRNA expression was observed in WiDr LF cells. These cells acquired approximately 2-fold resistance to mitoxantrone compared with the HF cell line, a phenotype that could be reverted by the BCRP inhibitor Ko143. Of note, WiDr cells expressed BCRP in the intracellular compartment, similarly to what we have described for Caco-2 cells. Our results provide further evidence for an important role of cellular folate status in the modulation of the expression of multidrug resistance transporters in cancer cells. We show that up-regulation of intracellularly localized BCRP in response to adaptation to LF conditions may be a common feature within a panel of colon cancer cell lines. Under these circumstances, folate supplementation might improve the efficacy of chemotherapeutic drugs by decreasing BCRP expression.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Folic Acid/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Genes, MDR , Neoplasm Proteins/genetics , Neoplasms/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/metabolism , Adaptation, Physiological/drug effects , Adaptation, Physiological/genetics , Antineoplastic Agents/pharmacology , Cell Line, Tumor/drug effects , Cell Line, Tumor/metabolism , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Female , Folic Acid/metabolism , Folic Acid/physiology , Humans , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Mitoxantrone/pharmacology , Multidrug Resistance-Associated Proteins/genetics , Multidrug Resistance-Associated Proteins/metabolism , Neoplasm Proteins/metabolism , Neoplasms/metabolism , Peptide Synthases/genetics , Peptide Synthases/metabolism , Tetrahydrofolate Dehydrogenase/genetics , Tetrahydrofolate Dehydrogenase/metabolism , Tissue Distribution
20.
J Steroid Biochem Mol Biol ; 113(3-5): 163-70, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19103290

ABSTRACT

8-Prenylnaringenin (8PN), one of the strongest plant-derived oestrogen receptors (ERs) ligand, has been suggested to have potential cancer chemo-preventive activities and anti-angiogenic properties. Because published data suggest that ERs serve as nodal point that allows interactions between hormones and growth factors mediated pathways, we decided to investigate the effects exerted by 8PN on Epidermal growth factor (EGF)-elicited pathways in breast cancer cells. Here we show that in ER positive MCF-7 cells, 8PN interferes with EGF induced cell proliferation by strongly inhibiting activation of PI(3)K/Akt pathway, without affecting EGFR expression or tyrosine phosphorylation, and exerting a synergistic activation of Erk1/2 phosphorylation. Moreover, we demonstrate that 8PN is a direct inhibitor of PI(3)K activity as it is shown by in vitro experiments with the purified enzyme and by its inability to impair serine phosphorylation of a constitutive active form of Akt. These findings suggest that inhibition of PI(3)K is a novel mechanism which contributes to 8PN activity to inhibit cancer cell survival and EGF induced proliferation.


Subject(s)
Breast Neoplasms/metabolism , Cell Line, Tumor , Epidermal Growth Factor/pharmacology , Flavanones/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phytoestrogens/metabolism , Cell Line, Tumor/drug effects , Cell Line, Tumor/metabolism , Cyclin D1/metabolism , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Humans , Molecular Structure , Receptors, Estrogen/metabolism , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL