Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Anticancer Res ; 40(6): 3191-3201, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32487613

ABSTRACT

BACKGROUND/AIM: Although it has been accepted that the tandem repeat galectin-8 (Gal-8) is linked to angiogenesis, the underlying mechanisms in endothelial cells has remained poorly understood. In this study we aimed to investigate the effect of Gal-8 on selected biological processes linked to angiogenesis in in vitro and in vivo models. MATERIALS AND METHODS: In detail, we assessed how exogenously added human recombinant Gal-8 (with or without vascular endothelial growth factor - VEGF) affects selected steps involved in vessel formation in human umbilical vein endothelial cells (HUVECs) as well as using the chick chorioallantoic membrane (CAM) assay. Gene expression profiling of HUVECs was performed to extend the scope of our investigation. RESULTS: Our findings demonstrate that Gal-8 in combination with VEGF enhanced cell proliferation and migration, two cellular events linked to angiogenesis. However, Gal-8 alone did not exhibit any significant effects on cell proliferation or on cell migration. The molecular analysis revealed that Gal-8 in the presence of VEGF influenced cytokine-cytokine receptor interactions, HIF-1 and PI3K/AKT signaling pathways. Gal-8 alone also targeted cytokine-cytokine receptor interactions, but with a different expression profile as well as a modulated focal adhesion and TNF signaling. CONCLUSION: Gal-8 promotes a pro-angiogenic phenotype possibly in a synergistic manner with VEGF.


Subject(s)
Chorioallantoic Membrane/blood supply , Chorioallantoic Membrane/drug effects , Galectins/pharmacology , Human Umbilical Vein Endothelial Cells/drug effects , Vascular Endothelial Growth Factor A/metabolism , Animals , Cell Movement/drug effects , Chick Embryo , Chorioallantoic Membrane/metabolism , Galectins/metabolism , Gene Expression Profiling , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , In Vitro Techniques , Neovascularization, Physiologic/drug effects
2.
Molecules ; 23(7)2018 Jul 04.
Article in English | MEDLINE | ID: mdl-29973576

ABSTRACT

Selective estrogen receptor modulators (SERMs) have been developed to achieve beneficial effects of estrogens while minimizing their side effects. In this context, we decided to evaluate the protective effect of genistein, a natural SERM, on skin flap viability in rats and in a series of in vitro experiments on endothelial cells (migration, proliferation, antioxidant properties, and gene expression profiling following genistein treatment). Our results showed that administration of genistein increased skin flap viability, but importantly, the difference is only significant when treatment is started 3 days prior the flap surgery. Based on our in vitro experiments, it may be hypothesized that the underlying mechanism may rather by mediated by increasing SOD activity and Bcl-2 expression. The gene expression profiling further revealed 9 up-regulated genes (angiogenesis/inflammation promoting: CTGF, CXCL5, IL-6, ITGB3, MMP-14, and VEGF-A; angiogenesis inhibiting: COL18A1, TIMP-2, and TIMP-3). In conclusion, we observed a protective effect of genistein on skin flap viability which could be potentially applied in plastic surgery to women undergoing a reconstructive and/or plastic intervention. Nevertheless, further research is needed to explain the exact underlying mechanism and to find the optimal treatment protocol.


Subject(s)
Endothelial Cells/cytology , Genistein/administration & dosage , Proto-Oncogene Proteins c-bcl-2/metabolism , Superoxide Dismutase/metabolism , Surgical Flaps/physiology , Animals , Cell Survival , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Gene Expression Profiling , Gene Expression Regulation/drug effects , Genistein/pharmacology , Human Umbilical Vein Endothelial Cells , Humans , Models, Animal , Rats , Time Factors , Up-Regulation
3.
Molecules ; 23(1)2018 Jan 17.
Article in English | MEDLINE | ID: mdl-29342121

ABSTRACT

In the present study we evaluated the anti-angiogenic activities of ß-escin (the major active compound of Aesculus hippocastanum L. seeds). Human umbilical-vein endothelial cells (HUVECs) were used as an in vitro model for studying the molecular mechanism underlying the anti-angiogenic effect of ß-escin. We investigated the in vitro effects on proliferation, migration, and tube formation of HUVECs and in vivo anti-angiogenic activity was evaluated in a chick chorioallantoic membrane (CAM) angiogenesis assay. Moreover, the effect on gene expressions was determined by the RT2 ProfilerTM human angiogenesis PCR Array. It was found that ß-escin exerts inhibitory effect on the basic fibroblast growth factor (bFGF)-induced proliferation, migration and tube formation, as well as CAM angiogenesis in vivo. The inhibition of critical steps of angiogenic process observed with ß-escin could be partially explained by suppression of Akt activation in response to bFGF. Moreover, the anti-angiogenic effects of ß-escin could also be mediated via inhibition of EFNB2 and FGF-1 gene expressions in endothelial cells. In conclusion, ß-escin affects endothelial cells as a negative mediator of angiogenesis in vitro and in vivo and may therefore be considered as a promising candidate for further research elucidating its underlying mechanism of action.


Subject(s)
Escin/chemistry , Escin/pharmacology , Human Umbilical Vein Endothelial Cells/drug effects , Neovascularization, Physiologic/drug effects , Cell Adhesion Molecules/metabolism , Cell Cycle/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Chromatography, High Pressure Liquid , Fibroblast Growth Factor 2/metabolism , Gene Expression Regulation/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Mass Spectrometry , Signal Transduction/drug effects , Transcriptome
4.
Int J Mol Sci ; 19(1)2017 Dec 22.
Article in English | MEDLINE | ID: mdl-29271940

ABSTRACT

Chalcones are precursors of flavonoid biosynthesis in plants. Both flavonoids and chalcones are intensively investigated because of a large spectrum of their biological activities. Among others, anticancer and antiangiogenic effects account for the research interest of these substances. Because of an essential role in cancer growth and metastasis, angiogenesis is considered to be a promising target for cancer treatment. Currently used antiangiogenic agents are either synthetic compounds or monoclonal antibodies. However, there are some limitations of their use including toxicity and high price, making the search for new antiangiogenic compounds very attractive. Nowadays it is well known that several natural compounds may modulate basic steps in angiogenesis. A lot of studies, also from our lab, showed that phytochemicals, including polyphenols, are potent modulators of angiogenesis. This review paper is focused on the antiangiogenic effect of flavonoids and chalcones and discusses possible underlying cellular and molecular mechanisms.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Chalcones/pharmacology , Flavonoids/pharmacology , Neoplasms/blood supply , Neovascularization, Pathologic/drug therapy , Angiogenesis Inhibitors/chemistry , Angiogenesis Inhibitors/therapeutic use , Animals , Chalcones/chemistry , Chalcones/therapeutic use , Flavonoids/chemistry , Flavonoids/therapeutic use , Humans , Hypoxia-Inducible Factor 1/metabolism , Neoplasms/complications , Neoplasms/drug therapy , Neoplasms/metabolism , Neovascularization, Pathologic/complications , Neovascularization, Pathologic/metabolism , Signal Transduction/drug effects , Vascular Endothelial Growth Factor A/metabolism
5.
Molecules ; 22(11)2017 Oct 26.
Article in English | MEDLINE | ID: mdl-29072623

ABSTRACT

It is now suggested that the inhibition of biological programs that are associated with the tumor microenvironment may be critical to the diagnostics, prevention and treatment of cancer. On the other hand, a suitable wound microenvironment would accelerate tissue repair and prevent extensive scar formation. In the present review paper, we define key signaling molecules (growth factors, cytokines, chemokines, and galectins) involved in the formation of the tumor microenvironment that decrease overall survival and increase drug resistance in cancer suffering patients. Additional attention will also be given to show whether targeted modulation of these regulators promote tissue regeneration and wound management. Whole-genome transcriptome profiling, in vitro and animal experiments revealed that interleukin 6, interleukin 8, chemokine (C-X-C motif) ligand 1, galectin-1, and selected proteins of the extracellular matrix (e.g., fibronectin) do have similar regulation during wound healing and tumor growth. Published data demonstrate remarkable similarities between the tumor and wound microenvironments. Therefore, tailor made manipulation of cancer stroma can have important therapeutic consequences. Moreover, better understanding of cancer cell-stroma interaction can help to improve wound healing by supporting granulation tissue formation and process of reepithelization of extensive and chronic wounds as well as prevention of hypertrophic scars and formation of keloids.


Subject(s)
Neoplasms/metabolism , Neoplasms/pathology , Signal Transduction , Tumor Microenvironment , Animals , Cellular Microenvironment , Cytokines/metabolism , Galectins/metabolism , Humans , Immune System/cytology , Immune System/immunology , Immune System/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Keloid/metabolism , Keloid/pathology , Neoplasms/immunology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Wound Healing , Wounds and Injuries/immunology , Wounds and Injuries/metabolism , Wounds and Injuries/pathology
6.
Biomed Pharmacother ; 89: 578-590, 2017 May.
Article in English | MEDLINE | ID: mdl-28258040

ABSTRACT

The formation of new blood vessels plays a crucial for the development and progression of pathophysiological changes associated with a variety of disorders, including carcinogenesis. Angiogenesis inhibitors (anti-angiogenics) are an important part of treatment for some types of cancer. Some natural products isolated from marine invertebrates have revealed antiangiogenic activities, which are diverse in structure and mechanisms of action. Many preclinical studies have generated new models for further modification and optimization of anti-angiogenic substances, and new information for mechanistic studies and new anti-cancer drug candidates for clinical practice. Moreover, in the last decade it has become apparent that galectins are important regulators of tumor angiogenesis, as well as microRNA. MicroRNAs have been validated to modulate endothelial cell migration or endothelial tube organization. In the present review we summarize the current knowledge regarding the role of marine-derived natural products, galectins and microRNAs in tumor angiogenesis.


Subject(s)
Angiogenesis Modulating Agents/pharmacology , Antineoplastic Agents/pharmacology , Neoplasms/drug therapy , Angiogenesis Inhibitors/pharmacology , Animals , Biological Products/pharmacology , Galectins/drug effects , Humans , Marine Toxins/pharmacology , MicroRNAs/drug effects
7.
Biomed Pharmacother ; 85: 463-471, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27887849

ABSTRACT

Ovarian carcinoma is initially sensitive to platinum-based therapy, but become resistant over time. The study of cancer sensitizing substance is therefore the major challenge for a number of scientific groups. Our experiments were carried out on human ovarian adenocarcinoma A2780cis cells resistant to cisplatin and their response to 2-(4'fluoro-phenylamino)-4H-1,3-thiazine[6,5-b]indole (thiazine[6,5-b]indole) and/or heat shock protein (Hsp) 90 inhibitor 17-dimethylaminoethylamino-17-demethoxygeldanamycin (17-DMAG) using proliferation assay, cell cycle analysis and monitoring of apoptosis were examined. A2780cis cells revealed the same fold of resistance to Hsp90 inhibitor 17-DMAG as it is declared for cisplatin (18 times), but only 3.2 times for thiazine[6,5-b]indole. Our results showed that the combination of thiazine[6,5-b]indole and 17-DMAG significantly reduced proliferation of A2780cis cells and led to their accumulation in G2/M phase of the cell cycle. Moreover, both thiazine[6,5-b]indole as well as 17-DMAG increased the number of annexin V positive A2780cis cells in time dependent manner. Interestingly, thiazine[6,5-b]indole treatment significantly activated also caspase-3 compared to untreated or 17-DMAG-treated cells and reduced mitochondrial membrane potential (MMP) of A2780cis cells with more significant decline after combined treatment. In this regard, the incubation of A2780cis cells with thiazine[6,5-b]indole induced PARP protein cleavage as well as an increased level of Bad protein with more pronounced changes after combined treatment. Importantly, Hsp70 protein was not upregulated in A2780cis cells neither by individual treatment nor by mutual combination. Our results signify antiproliferative and pro-apoptotic effects of novel thiazine[6,5-b]indole potentiated by Hsp90 inhibitor 17-DMAG in ovarian adenocarcinoma cells resistant to cisplatin and therefore represents new strategy in cancer treatment.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Agents/pharmacology , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Indoles/pharmacology , Ovarian Neoplasms/drug therapy , Thiazines/pharmacology , Antineoplastic Agents/chemistry , Benzoquinones/pharmacology , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cisplatin/pharmacology , Dose-Response Relationship, Drug , Female , HSP90 Heat-Shock Proteins/metabolism , Humans , Indoles/chemistry , Lactams, Macrocyclic/pharmacology , Thiazines/chemistry
8.
Int J Mol Med ; 37(1): 21-8, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26397183

ABSTRACT

Estrogen deprivation is considered responsible for many age-related processes, including poor wound healing. Guided by previous observations that estradiol accelerates re­epithelialization through estrogen receptor (ER)­ß, in the present study, we examined whether selective ER agonists [4,4',4''-(4-propyl [1H] pyrazole-1,3,5-triyl)­trisphenol (PPT), ER­α agonist; 2,3-bis(4-hydroxyphenyl)-propionitrile (DPN), ER­ß agonist] affect the expression of basic proliferation and differentiation markers (Ki­67, keratin­10, ­14 and ­19, galectin­1 and Sox­2) of keratinocytes using HaCaT cells. In parallel, ovariectomized rats were treated daily with an ER modulator, and wound tissue was removed 21 days after wounding and routinely processed for basic histological analysis. Our results revealed that the HaCaT keratinocytes expressed both ER­α and ­ß, and thus are well-suited for studying the effects of ER agonists on epidermal regeneration. The activation of ER­α produced a protein expression pattern similar to that observed in the control culture, with a moderate expression of Ki­67 being observed. However, the activation of ER­ß led to an increase in cell proliferation and keratin­19 expression, as well as a decrease in galectin­1 expression. Fittingly, in rat wounds treated with the ER­ß agonist (DPN), epidermal regeneration was accelerated. In the present study, we provide information on the mechanisms through which estrogens affect the expression patterns of selected markers, thus modulating keratinocyte proliferation and differentiation; in addition, we demonstrate that the pharmacological activation of ER-α and -ß has a direct impact on wound healing.


Subject(s)
Estrogen Receptor alpha/agonists , Estrogen Receptor beta/agonists , Keratinocytes/drug effects , Nitriles/pharmacology , Phenols/pharmacology , Pyrazoles/pharmacology , Wound Healing/drug effects , Animals , Cell Differentiation/drug effects , Cell Line , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Female , Humans , Keratinocytes/cytology , Keratinocytes/metabolism , Keratinocytes/pathology , Rats, Sprague-Dawley , Skin/drug effects , Skin/metabolism , Skin/pathology
9.
Molecules ; 20(11): 20538-50, 2015 Nov 18.
Article in English | MEDLINE | ID: mdl-26593896

ABSTRACT

Reactive oxygen species (ROS) are highly considered in the ethiopathogenesis of different pathological conditions because they may cause significant damage to cells and tissues. In this paper, we focused on potential antioxidant properties of two medical plants such as the Agrimonia eupatoria L. and Cynara cardunculus L. Both plants have previously been studied for their pharmacological activities, especially as hepatoprotective and hypoglycemic activities. It has been suggested, that their effects are related to the antioxidant properties of polyphenols, which are dominant compounds of the plants' extracts. In the present study HPLC-MS analysis of water infusion was performed allowing the identification of several phenolic constituents. Furthermore, antioxidant effects of the two extracts were compared showing higher effects for agrimony extract compared to artichoke. Thus, agrimony was selected for the in vivo study using the skin flap viability model. In conclusion, our results provide evidence that the A. eupatoria extract may be a valuable source of polyphenols to be studied for the future development of supplements useful in the prevention of diseases linked to oxidative stress.


Subject(s)
Agrimonia/chemistry , Antioxidants/chemistry , Cynara/chemistry , Phenols/chemistry , Plant Extracts/chemistry , Animals , Antioxidants/pharmacology , Catalase/metabolism , Cell Line/drug effects , Cell Line/metabolism , Cell Survival/drug effects , Chromatography, High Pressure Liquid , DNA Damage/drug effects , Humans , Oxidative Stress/drug effects , Phenols/pharmacology , Plant Extracts/pharmacology , Rats , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism
10.
Int J Mol Sci ; 16(5): 11728-49, 2015 May 22.
Article in English | MEDLINE | ID: mdl-26006245

ABSTRACT

Epidemiological studies have revealed that high consumption of soy products is associated with low incidences of hormone-dependent cancers, including breast and prostate cancer. Soybeans contain large amounts of isoflavones, such as the genistein and daidzain. Previously, it has been demonstrated that genistein, one of the predominant soy isoflavones, can inhibit several steps involved in carcinogenesis. It is suggested that genistein possesses pleiotropic molecular mechanisms of action including inhibition of tyrosine kinases, DNA topoisomerase II, 5α-reductase, galectin-induced G2/M arrest, protein histidine kinase, and cyclin-dependent kinases, modulation of different signaling pathways associated with the growth of cancer cells (e.g., NF-κB, Akt, MAPK), etc. Moreover, genistein is also a potent inhibitor of angiogenesis. Uncontrolled angiogenesis is considered as a key step in cancer growth, invasion, and metastasis. Genistein was found to inhibit angiogenesis through regulation of multiple pathways, such as regulation of VEGF, MMPs, EGFR expressions and NF-κB, PI3-K/Akt, ERK1/2 signaling pathways, thereby causing strong antiangiogenic effects. This review focuses on the antiangiogenic properties of soy isoflavonoids and examines their possible underlying mechanisms.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Breast Neoplasms/blood supply , Breast/blood supply , Glycine max/chemistry , Isoflavones/pharmacology , Neovascularization, Pathologic/drug therapy , Signal Transduction/drug effects , Angiogenesis Inhibitors/chemistry , Angiogenesis Inhibitors/therapeutic use , Animals , Breast/drug effects , Breast/metabolism , Breast/pathology , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Female , Genistein/chemistry , Genistein/pharmacology , Genistein/therapeutic use , Humans , Isoflavones/chemistry , Isoflavones/therapeutic use , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology
11.
Cas Lek Cesk ; 153(5): 231-7, 2014.
Article in Czech | MEDLINE | ID: mdl-25370768

ABSTRACT

Galectins are representatives of endogenous lectins - molecules specifically recognizing distinct sugar motifs. They play an important role in the processes of cell proliferation, differentiation, migration and extracellular matrix formation. Furthermore, galectins are able to transfer cellular signals and to participate in intercellular interaction. It has been proven that galectins play an important role in the formation of tumor and/or wound healing microenvironment. This review contains an overview of experimental and clinical studies dealing with biological roles of galectins in tissue repair and in its parallel - the tumor growth.


Subject(s)
Galectins/physiology , Neoplasms/physiopathology , Wound Healing/physiology , Animals , Cell Transformation, Neoplastic/pathology , Disease Progression , Extracellular Matrix/pathology , Extracellular Matrix/physiology , Humans , Neoplasms/pathology
12.
Anticancer Res ; 34(8): 3991-6, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25075021

ABSTRACT

BACKGROUND/AIM: Stromal cells in the tumor microenvironment are primarily considered as sources of promalignant factors. The objective of our study was to define the effect of extracellular matrix (ECM) produced by normal dermal or cancer-associated fibroblasts exposed to adhesion/growth-regulatory lectin galectin-1 on human umbilical vein endothelial cells (HUVECs). MATERIALS AND METHODS: Fibroblasts were cultured for 10 days with lectin, followed by removing cellular constituents after an osmotic shock. Freshly-isolated HUVECs were placed on the ECM. In parallel, HUVECs were seeded on untreated and gelatin-coated surfaces as controls. A positive control for growth of HUVECs culture using medium supplemented with vascular endothelial growth factor completed the test panel. Cells were kept in contact to the substratum for two days and then processed for immunocytochemistry. RESULTS: HUVECs seeded on fibroblast-generated ECM presented a comparatively high degree of proliferation. Furthermore, contact to substratum produced by tumor-associated fibroblasts led to generation of a meshwork especially rich in fibronectin. CONCLUSION: Galectin-1 is apparently capable to trigger ECM production favorable for growth of HUVECs, prompting further work on characterizing structural features of the ECM and in situ correlation of lectin presence, ECM constitution and neoangiogenesis.


Subject(s)
Extracellular Matrix/metabolism , Fibroblasts/physiology , Galectin 1/pharmacology , Tumor Microenvironment , Cell Proliferation/drug effects , Cells, Cultured , Human Umbilical Vein Endothelial Cells/physiology , Humans , Vascular Endothelial Growth Factor A/pharmacology
13.
Bioorg Med Chem ; 21(21): 6623-33, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-24012378

ABSTRACT

An effective synthesis of analogs of the indole phytoalexin cyclobrassinin with NR1R2 group instead of SCH3 was developed starting from indole-3-carboxaldehyde. The target compounds were prepared by spirocyclization of 1-Boc-thioureas with the formation of isolable spiroindoline intermediates, followed by the trifluoroacetic acid-induced cascade reaction consisting of methanol elimination, deprotection and rearrangement of the iminium ion. The structures of novel products were elucided by the (1)H and (13)C NMR spectroscopy, including HMBC, HSQC, COSY, NOESY and DEPT measurements. Several newly synthesized compounds demonstrated significant antiproliferative/cytotoxic activity against human leukemia and solid tumor cell lines, as well as remarkable selectivity of these effects against cancer cells relative to the non-malignant HUVEC cells.


Subject(s)
Antineoplastic Agents/chemical synthesis , Indoles/chemistry , Sesquiterpenes/chemistry , Thiocarbamates/chemistry , Antineoplastic Agents/chemistry , Antineoplastic Agents/toxicity , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Screening Assays, Antitumor , HeLa Cells , Human Umbilical Vein Endothelial Cells , Humans , Indoles/chemical synthesis , Indoles/toxicity , Jurkat Cells , MCF-7 Cells , Magnetic Resonance Spectroscopy , Stereoisomerism , Thiocarbamates/chemical synthesis , Thiocarbamates/toxicity , Phytoalexins
14.
Eur J Med Chem ; 66: 46-55, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23792315

ABSTRACT

A series of alkylphosphocholine and alkylphosphohomocholine derivatives of cetyltrimethylammonium bromide, cetylpyridinium bromide, benzalkonium bromide (C16) and benzethonium chloride have been synthesized. Their physicochemical properties were also investigated. The critical micelle concentration (cmc), the surface tension value at the cmc (γcmc), and the surface area at the surface saturation per head group (Acmc) were determined by means of surface tension measurements. The prepared compounds exhibit significant cytotoxic, antifungal and antiprotozoal activities. Alkylphosphocholines and alkylphosphohomocholines possess higher antifungal activity against Candida albicans in comparison with quaternary ammonium compounds in general. However, quaternary ammonium compounds exhibit significantly higher activity against human tumor cells and pathogenic free-living amoebae Acanthamoeba lugdunensis and Acanthamoeba quina compared to alkylphosphocholines. The relationship between structure, physicochemical properties and biological activity of the tested compounds is discussed.


Subject(s)
Benzalkonium Compounds/chemistry , Benzethonium/chemistry , Cetrimonium Compounds/chemistry , Cetylpyridinium/chemistry , Phosphorylcholine/chemical synthesis , Phosphorylcholine/pharmacology , Antifungal Agents/chemical synthesis , Antifungal Agents/chemistry , Antifungal Agents/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Candida albicans/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cetrimonium , Chemistry Techniques, Synthetic , Humans , Micelles , Phosphorylcholine/chemistry , Structure-Activity Relationship , Surface Properties
15.
Oncol Rep ; 30(3): 1455-62, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23807540

ABSTRACT

Erythropoietin (Epo) is a key regulator of erythroid cell proliferation, differentiation and apoptosis. In the form of the recombinant protein, it is widely used to treat various types of anemias, including that associated with cancer and with the myelosuppressive effects of chemotherapy, particularly platinum-based regimens. Our previous studies confirmed the presence of Epo receptors (EpoRs) in ovarian adenocarcinoma cell lines and demonstrated that long-term Epo treatment of A2780 cells resulted in the development of a phenotype exhibiting both enhanced Epo signaling and increased paclitaxel resistance. In the present study, we carried out a series of experiments to analyze the pro-angiogenic potential of Epo-treated A2780 and SKOV-3 cells. Our studies revealed that conditioned media of Epo-treated A2780 cells had a stimulative effect on human umbilical vein endothelial cells (HUVECs). This effect was only seen when A2780 cells were incubated under hypoxic conditions. Furthermore, Epo increased the secretion of interleukin (IL)-4, IL-5, IL-6, IL-8, IL-10, IL-12, IL-13, GM-CSF and interferon-γ by A2780 cells that grew in hypoxic conditions. In this regard, conditioned media of hypoxic and Epo-treated A2780 cells induced a significant phosphorylation of STAT-5 in HUVECs. Our results may have important implications for ovarian cancer patients receiving Epo.


Subject(s)
Adenocarcinoma/blood supply , Cell Proliferation , Erythropoietin/pharmacology , Hypoxia/physiopathology , Neovascularization, Pathologic , Ovarian Neoplasms/blood supply , Adenocarcinoma/drug therapy , Adenocarcinoma/pathology , Blotting, Western , Culture Media, Conditioned/pharmacology , Enzyme-Linked Immunosorbent Assay , Epoetin Alfa , Female , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Recombinant Proteins/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
16.
Mol Biol Rep ; 40(7): 4571-80, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23666054

ABSTRACT

In the present investigation a novel series of chalcone analogues were synthesized and evaluated for their anti-proliferative activity in human umbilical vein endothelial cells (HUVECs). Among 14 tested compounds, chalcone analogue (E)-3-(2'-methoxybenzylidene)-4-chromanone (KRP6) exhibited the most potent activity with IC50 19 µM. Moreover, HUVECs exhibited divergent, even opposing concentration-dependent responses to KRP6. This compound was the most potent inhibitor of cell proliferation and extracellular matrix formation (fibronectin and type IV collagen) at higher concentrations (20-50 µM). In contrast, KRP6 stimulated the compensatory increase in proliferative activity including extracellular matrix formation at low concentrations (1, 10 µM). KRP6 concentration-dependently modulated phosphorylation of Akt and mitogen-activated protein kinases such as extracellular signal-regulated kinase-1/-2 and p38 kinase, suggesting that these pathways play a role in the effect mediated by this compound. In addition, we found a selective effect on activated endothelial cells, in particular with resting endothelial cells. In conclusion, KRP6 is a potent modulator of selected steps of the angiogenic process in vitro. Accordingly, further in vivo research should be performed to facilitate its use in clinical practice.


Subject(s)
Chalcone/pharmacology , Human Umbilical Vein Endothelial Cells/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Chalcone/analogs & derivatives , Chalcone/chemistry , Extracellular Matrix/metabolism , Humans , Signal Transduction/drug effects
17.
Toxicol In Vitro ; 27(2): 939-44, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23339777

ABSTRACT

The aim of the study was to investigate the cytotoxic activity of camalexin and its five synthetic derivatives in cancer and non-cancer cells. In cancer cells the benzocamalexin (BC) displayed the most potent activity with an IC value of 23.3-30.1 µmol/L. On the other hand, minimal toxicity (IC>100.0 µmol/L) in non-cancer cells was observed. Based on these results, BC was selected for further studies. Flow cytometric analysis revealed a BC-induced arrest of the cell cycle in the G2 phase associated with downregulation of α-tubulin, α1-tubulin, ß5-tubulin expression. These findings suggest that the inhibitory effect of BC is mediated via inhibition of microtubule formation. Moreover, BC downregulated the expression of microtubule-related protein indicating the effect of this compound on microtubule assembly. After treatment with BC increase of the sub-G DNA content fraction was noted which is considered to be a marker of apoptotic cell death. Apoptosis was also confirmed by DNA fragmentation assay. Moreover, quantitative real-time PCR showed that BC downregulated the expression of antiapoptotic genes Bcl-2 and Bcl-xL and upregulated the expression of proapoptotic Bax. Taken together, our study suggests that the blockade of cell cycle progression and initiation of apoptosis may play an important role in the antiproliferative activity of BC in human cancer cells.


Subject(s)
Anti-Infective Agents/toxicity , Antineoplastic Agents, Phytogenic/toxicity , Indoles/toxicity , Thiazoles/toxicity , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , DNA Fragmentation , Human Umbilical Vein Endothelial Cells , Humans , Proto-Oncogene Proteins c-bcl-2/genetics , RNA, Messenger/metabolism , Tubulin/genetics , bcl-2-Associated X Protein/genetics
18.
Phytother Res ; 27(2): 159-65, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22451355

ABSTRACT

This study was designed to examine the in vitro antiproliferative effect of the horse chestnut extract (HCE) on cancer cell lines. Furthermore, we have investigated the in vitro effect of HCE on some angiogenic events by using human umbilical vein endothelial cells. The cell proliferation was evaluated by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay and anchorage-independent growth by colony-forming assay. To understand the growth inhibitory effects, carcinoma cell lines (Jurkat, CEM, HeLa, and MCF-7) were treated with various concentrations of HCE. Incubation of Jurkat, CEM, HeLa, and MCF-7 cancer cells with HCE at 125 µg/mL for 72 h caused 93.7%, 32.3%, 20.4% and 40.4% reduction in cell survival. Colony-forming assay also confirmed growth-inhibitory effects of the compound studied. In HeLa HCE-treated cells, we found a significant increase in cells having sub-G(0) /G(1) DNA content which is considered to be a marker of apoptotic cell death. Apoptosis was also further confirmed by DNA fragmentation analysis.Furthermore, HCE inhibited migration of human umbilical vein endothelial cells as well as decreased secretion of matrix metalloproteinase and vascular endothelial growth factor.In conclusion, the present study has assessed the in vitro antiproliferative/antiangiogenic potential of HCE. These results generate a rationale for in vivo efficacy studies with horse chestnut in preclinical cancer models.


Subject(s)
Aesculus/chemistry , Angiogenesis Inhibitors/pharmacology , Cell Proliferation/drug effects , Plant Extracts/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Movement/drug effects , DNA Fragmentation/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Vascular Endothelial Growth Factor A/metabolism
19.
Eur J Pharmacol ; 691(1-3): 125-33, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22721615

ABSTRACT

Angiogenesis, the growth of new blood vessels, is necessary for cancerous tumors to keep growing and spreading. Suppression of abnormal angiogenesis may provide therapeutic strategies for the treatment of angiogenesis-dependent disorders. In the present study, we describe the in vitro and in vivo anti-angiogenic activities of the flavonoid precursor 4-hydroxychalcone (Q797). This chalcone (22µg/ml) suppressed several steps of angiogenesis, including endothelial cell proliferation, migration and tube formation without showing any signs of cytotoxicity. Moreover, we found a selective effect on activated endothelial cells, in particular with resting endothelial cells and the human epithelial tumor cell lines (HeLa, MCF-7, A549). In addition, Q797 was able to modulate both vascular endothelial growth factor (VEGF)- and basic fibroblast growth factor (FGF)- induced phosphorylation of extracellular signal-regulated kinase (ERK)-1/-2 and Akt kinase. It did not influence the nuclear translocation of p65 subunit of the nuclear factor-κB (NF-κB) when human endothelial cells were stimulated with tumor necrosis factor (TNF)-α. Taken together this indicates that the Q797-mediated inhibition of in vitro angiogenic features of endothelial cells is most likely caused by suppression of growth factor pathways. The potent inhibitory effect of Q797 on bFGF-driven neovascularization was also demonstrated in vivo using the chick chorioallantoic membrane (CAM) assay. In summary, this chalcone could serve as a new leading structure in the discovery of new potent synthetic angiogenesis inhibitors.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Chalcones/pharmacology , Angiogenesis Inhibitors/chemistry , Angiogenesis Inhibitors/therapeutic use , Animals , Capillaries/drug effects , Capillaries/physiology , Cell Movement/drug effects , Cell Proliferation/drug effects , Chalcones/chemistry , Chalcones/therapeutic use , Chick Embryo , Drug Design , Endothelial Cells/cytology , Endothelial Cells/drug effects , Fibrin/metabolism , HeLa Cells , Humans , Intracellular Space/drug effects , Intracellular Space/metabolism , Neovascularization, Pathologic/drug therapy , Signal Transduction/drug effects
20.
Oncol Rep ; 28(1): 141-6, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22552716

ABSTRACT

Erythropoietin (Epo) is a critical regulator of erythroid cell proliferation, differentiation and apoptosis. In the form of a recombinant protein, it is widely used to treat various forms of anemia, including that associated with cancer and with the myelosuppressive effects of chemotherapy. Studies of ovarian cancer cell lines have demonstrated the presence of the Epo receptor (EpoR), but there are disagreements regarding its localization and functionality in these cells. Using fluorescence microscopy, we were not able to identify the EpoR on the surface of A2780 cells, in contrast to the positive control K562 cells. Flow cytometry did reveal a weak surface EpoR signal in A2780 cells. Interestingly, most of the EpoR in A2780 cells was found in the cytoplasm, more abundantly as an intracellular membrane-associated protein than a soluble one. Silencing EpoR expression by lentiviral-mediated shRNA resulted in reduced A2780 proliferation as well as reduction in Epo-induced phosphorylation of Erk1/2. Our findings provide important insights into the biology of the EpoR in ovarian cancer cells.


Subject(s)
Receptors, Erythropoietin/physiology , Cell Line, Tumor , Cell Membrane/metabolism , Cell Proliferation , Endoplasmic Reticulum/metabolism , Erythropoietin/physiology , Female , Gene Knockdown Techniques , Humans , Intracellular Membranes/metabolism , Microscopy, Fluorescence , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Ovarian Neoplasms , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , Receptors, Erythropoietin/genetics , Receptors, Erythropoietin/metabolism , Secretory Vesicles/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...