Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS One ; 8(1): e54261, 2013.
Article in English | MEDLINE | ID: mdl-23349840

ABSTRACT

Cellular functions are regulated by complex networks of many different signaling pathways. The TGFß and cAMP pathways are of particular importance in tumor progression. We analyzed the cross-talk between these pathways in breast cancer cells in 2D and 3D cultures. We found that cAMP potentiated TGFß-dependent gene expression by enhancing Smad3 phosphorylation. Higher levels of total Smad3, as observed in 3D-cultured cells, blocked this effect. Two Smad3 regulating proteins, YAP (Yes-associated protein) and TßRI (TGFß receptor 1), were responsive to cAMP. While YAP had little effect on TGFß-dependent expression and Smad3 phosphorylation, a constitutively active form of TßRI mimicked the cAMP effect on TGFß signaling. In 3D-cultured cells, which show much higher levels of TßRI and cAMP, TßRI was unresponsive to cAMP. Upregulation of TßRI expression by cAMP was dependent on transcription. A proximal TßRI promoter fragment was moderately, but significantly activated by cAMP suggesting that cAMP increases TßRI expression at least partially by activating TßRI transcription. Neither the cAMP-responsive element binding protein (CREB) nor the TßRI-regulating transcription factor Six1 was required for the cAMP effect. An inhibitor of histone deacetylases alone or together with cAMP increased TßRI expression by a similar extent as cAMP alone suggesting that cAMP may exert its effect by interfering with histone acetylation. Along with an additive stimulatory effect of cAMP and TGFß on p21 expression an additive inhibitory effect of these agents on proliferation was observed. Finally, we show that mesenchymal stem cells that interact with breast cancer cells can simultaneously activate the cAMP and TGFß pathways. In summary, these data suggest that combined effects of cAMP and TGFß, as e.g. induced by mesenchymal stem cells, involve the upregulation of TßRI expression on the transcriptional level, likely due to changes in histone acetylation. As a consequence, cancer cell functions such as proliferation are affected.


Subject(s)
Cyclic AMP/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Protein Serine-Threonine Kinases/genetics , Receptors, Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/pharmacology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Blotting, Western , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Culture Techniques , Cell Line, Tumor , Cell Proliferation/drug effects , Coculture Techniques , Colforsin/pharmacology , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Female , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/metabolism , RNA Interference , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Smad3 Protein/genetics , Smad3 Protein/metabolism , Transcription Factors , YAP-Signaling Proteins
2.
Cancer Lett ; 325(2): 165-74, 2012 Dec 28.
Article in English | MEDLINE | ID: mdl-22771536

ABSTRACT

We performed next generation sequencing- and microarray-based gene expression profiling of CD44(+)/CD24(-)/CD45(-) breast CSCs (cancer stem cells) isolated from primary ERα-positive breast cancer. By combining semi-automated dissociation of human tumor tissue, magnetic cell sorting and cDNA amplification less than 500 CSCs were required for transcriptome analyses. Besides overexpressing genes involved in maintenance of stemness, the CSCs showed higher levels of genes that drive the PI3K pathway, including EGFR, HB-EGF, PDGFRA/B, PDGF, MET, PIK3CA, PIK3R1 and PIK3R2. This suggests that, in CSCs of ERα-positive breast cancer, the PI3K pathway which is involved in endocrine resistance is hyperactive.


Subject(s)
Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/pathology , Estrogens , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Neoplasm Proteins/physiology , Neoplasms, Hormone-Dependent/pathology , Nucleic Acid Amplification Techniques/methods , Phosphatidylinositol 3-Kinases/physiology , Breast Neoplasms/enzymology , CD24 Antigen/analysis , Carcinoma, Ductal, Breast/enzymology , Estrogen Receptor alpha/analysis , Female , Humans , Hyaluronan Receptors/analysis , Immunomagnetic Separation , Immunophenotyping , Isoenzymes/physiology , Neoplasm Proteins/genetics , Neoplasms, Hormone-Dependent/enzymology , Neoplastic Stem Cells/chemistry , Neoplastic Stem Cells/enzymology , Neoplastic Stem Cells/pathology , RNA, Messenger/genetics , RNA, Neoplasm/genetics , Sensitivity and Specificity , Transcriptome
3.
Oncol Rep ; 26(5): 1121-5, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21811762

ABSTRACT

Several members of the Ets (E26 transformation specific) transcription factor family are involved in tumor progression, e.g. by activating matrix metalloproteases. Ets proteins share a unique DNA-binding domain, the Ets domain, which specifically recognizes GGAA/T-containing sequences common in many promoters. While the roles of quite a number of Ets proteins in carcinogenesis have been well established, little is known about the importance of the Ets protein Elf-1 (E74-like factor 1) in cancer. Herein, we analyzed the expression of Elf-1 in breast cancer. We found that, like T-cells, breast cancer cells express both the 80 and 98 kDa isoforms of the Elf-1 protein with the 98 kDa isoform only be present in the nucleus. Immunohistochemical analysis of 119 breast cancer biopsies showed anti-Elf-1 immunoreactivity exclusively in the nucleus. Elf-1 expression varied largely among the breast cancer samples showing a negative correlation with histological grading. However, no association of Elf-1 expression with clinical outcome was observed, even when sub-cohorts of patients who received either only adjuvant endocrine treatment or only chemotherapy were separately analyzed. These data suggest that Elf-1 may modulate breast cancer progression to some extent without having an impact on survival of breast cancer patients.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Nuclear Proteins/biosynthesis , Transcription Factors/biosynthesis , Animals , Breast Neoplasms/genetics , Cell Line, Tumor , Female , Humans , Immunohistochemistry , Mice , Middle Aged , Neoplasm Grading , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Treatment Outcome
4.
Int J Oncol ; 39(3): 689-96, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21667024

ABSTRACT

Stromal cells, such as mesenchymal stem cells (MSCs) and carcinoma-associated fibroblasts (CAFs), play a role in cancer progression. To analyze their ability to modulate drug response, we generated spheroids of MCF-7 or MDA-MB-231 breast cancer cells in the absence or presence of human (h)MSCs or hCAFs and tested the susceptibility of the breast cancer cells to three different kinase inhibitors (TKI258, RAD001 and RAF265) used in cancer therapy. While stromal cells did not affect the response of either breast cancer cell line to the PDGFR/FGFR/VEGFR inhibitor TKI258, they sensitized breast cancer cells to the mTOR inhibitor RAD001. In MCF-7 cells, this was accompanied by increased apoptosis. hMSCs and to a lesser extent hCAFs also enhanced the cytotoxic effect of RAF inhibitor RAF265 on MDA-MB-231 cells. Searching for the mechanism that underlies the effect of stromal cells on RAF265 response we found that stromal cells inhibited RAF265-induced increase in ERK1/2 phosphorylation, supported RAF265-dependent downregulation of PKCα (protein kinase Cα) and prevented RAF265-induced conversion of LC3B, a marker of autophagy. To mimic the changes in ERK1/2 phosphorylation and PKCα expression in response to the stromal cells, we treated cells with MEK1 inhibitor U0126 or PKCα inhibitor Gö6976, respectively. U0126, but not Gö6976, was as effective as hMSCs in sensitizing MDA-MB-231 cells to RAF265. This suggests that hMSCs and hCAFs increased the cytotoxic effect of RAF265 on MDA-MB-231 cells by downregulating ERK1/2 phosphorylation. In summary, this study shows that hMSCs are able to render breast cancer cells more susceptible to kinase inhibitors and that, to the most part, hCAFs to which hMSCs can differentiate are able to mimic the drug-sensitizing effects of hMSCs.


Subject(s)
Breast Neoplasms/pathology , Breast Neoplasms/therapy , Cell Communication/physiology , Fibroblasts/pathology , Mesenchymal Stem Cells/pathology , Protein Kinase Inhibitors/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/enzymology , Cell Line, Tumor , Cells, Cultured , Coculture Techniques , Everolimus , Female , Humans , Imidazoles/pharmacology , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation/drug effects , Pyridines/pharmacology , Sirolimus/analogs & derivatives , Sirolimus/pharmacology , Stromal Cells/pathology , TOR Serine-Threonine Kinases/antagonists & inhibitors , raf Kinases/antagonists & inhibitors
5.
Cancer Res ; 70(1): 277-87, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-20048078

ABSTRACT

Targets that could improve the treatment of brain tumors remain important to define. This study of a transformation-associated isoform of alpha2-macroglobulin (A2M*) and its interaction with the low-density lipoprotein receptor-related protein-1 (LRP1) suggests a new mechanism for abrogating the malignant potential of astrocytoma cells. LRP1 bound A2M* found to be associated with an inhibition of tumor cell proliferation, migration, invasion, spheroid formation, and anchorage-independent growth. Transcriptional studies implicated effects on the Wnt/beta-catenin signaling pathway. Notably, LRP1 antibodies could phenocopy the effects of A2M*. Our findings suggest a pathway of tumor suppression in astrocytoma that might be tractable to therapeutic exploitation.


Subject(s)
Astrocytoma/metabolism , LDL-Receptor Related Protein-Associated Protein/metabolism , Signal Transduction/physiology , alpha-Macroglobulins/metabolism , beta Catenin/metabolism , Blotting, Western , Cell Adhesion/physiology , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation , Gene Expression , Humans , Immunohistochemistry
6.
PLoS One ; 3(10): e3508, 2008.
Article in English | MEDLINE | ID: mdl-18946510

ABSTRACT

BACKGROUND: Glyoxalases (Glo1 and Glo2) are involved in the glycolytic pathway by detoxifying the reactive methylglyoxal (MGO) into D-lactate in a two-step reaction using glutathione (GSH) as cofactor. Inhibitors of glyoxalases are considered as anti-inflammatory and anti-carcinogenic agents. The recent finding that various polyphenols modulate Glo1 activity has prompted us to assess curcumin's potency as an Glo1 inhibitor. METHODOLOGY/PRINCIPAL FINDINGS: Cultures of whole blood cells and tumor cell lines (PC-3, JIM-1, MDA-MD 231 and 1321N1) were set up to investigate the effect of selected polyphenols, including curcumin, on the LPS-induced cytokine production (cytometric bead-based array), cell proliferation (WST-1 assay), cytosolic Glo1 and Glo2 enzymatic activity, apoptosis/necrosis (annexin V-FITC/propidium iodide staining; flow cytometric analysis) as well as GSH and ATP content. Results of enzyme kinetics revealed that curcumin, compared to the polyphenols quercetin, myricetin, kaempferol, luteolin and rutin, elicited a stronger competitive inhibitory effect on Glo1 (K(i) = 5.1+/-1.4 microM). Applying a whole blood assay, IC(50) values of pro-inflammatory cytokine release (TNF-alpha, IL-6, IL-8, IL-1beta) were found to be positively correlated with the K(i)-values of the aforementioned polyphenols. Moreover, whereas curcumin was found to hamper the growth of breast cancer (JIMT-1, MDA-MB-231), prostate cancer PC-3 and brain astrocytoma 1321N1 cells, no effect on growth or vitality of human primary hepatocytes was elucidated. Curcumin decreased D-lactate release by tumor cells, another clue for inhibition of intracellular Glo1. CONCLUSIONS/SIGNIFICANCE: The results described herein provide new insights into curcumin's biological activities as they indicate that inhibition of Glo1 by curcumin may result in non-tolerable levels of MGO and GSH, which, in turn, modulate various metabolic cellular pathways including depletion of cellular ATP and GSH content. This may account for curcumin's potency as an anti-inflammatory and anti-tumor agent. The findings support the use of curcumin as a potential therapeutic agent.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Curcumin/pharmacology , Lactoylglutathione Lyase/antagonists & inhibitors , Blood Cells/drug effects , Blood Cells/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Drug Evaluation, Preclinical , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Humans , Interleukin-1beta/metabolism , L-Lactate Dehydrogenase/metabolism , Lipopolysaccharides/pharmacology , Models, Biological , Neoplasms/pathology , Phenols/pharmacology , Polyphenols , Substrate Specificity
7.
Biochem Pharmacol ; 76(5): 631-44, 2008 Sep 01.
Article in English | MEDLINE | ID: mdl-18625205

ABSTRACT

Esters of alpha-oxo-carbonic acids such as ethyl pyruvate (EP) have been demonstrated to exert inhibitory effects on the production of anti-inflammatory cytokines. So far, there is no information about effects, if any, of ethyl lactate (EL), an obviously inactive analogue of EP, on inflammatory immune responses. In the present study, we provide evidence that the anti-inflammatory action of alpha-oxo-carbonic acid esters is mediated by inhibition of glyoxalases (Glo), cytosolic enzymes that catalyse the conversion of alpha-oxo-aldehydes such as methylglyoxal (MGO) into the corresponding alpha-hydroxy acids using glutathione as a cofactor. In vitro enzyme activity measurements revealed the inhibition of human Glo1 by alpha-oxo-carbonic acid esters, whilst alpha-hydroxy-carbonic acid esters such as EL were not inhibitory. In contrast, both EP and EL were shown to suppress the Lipopolysaccharide (LPS)-induced production of pro-inflammatory cytokines such as tumor necrosis factor-alpha, interleukin (IL)-1beta, IL-6 and IL-8 from human immunocompetent cells, and modulated the expression of the immune receptors HLA-DR, CD14 and CD91 on human monocytes. Here, we show a crossing link between glyoxalases and the immune system. The results described herein introduce glyoxalases as a possible target for therapeutic approaches of immune suppression.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cytokines/antagonists & inhibitors , Lactates/pharmacology , Lactoylglutathione Lyase/antagonists & inhibitors , Pyruvates/pharmacology , Receptors, Immunologic/metabolism , Animals , Cytokines/biosynthesis , Down-Regulation , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Macrophages/drug effects , Macrophages/enzymology , Mice , Monocytes/drug effects , Monocytes/enzymology , Pyruvaldehyde/pharmacology , Reverse Transcriptase Polymerase Chain Reaction
8.
Mol Pharmacol ; 70(6): 1892-901, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16971553

ABSTRACT

The extracellular part of transmembrane segment V (TM-V) is expected to be involved in the activation process of 7TM receptors, but its role is far from clear. Here, we study the highly constitutively active CXC-chemokine receptor encoded by human herpesvirus 8 (ORF74-HHV8), in which a metal ion site was introduced at the extracellular end of TM-V by substitution of two arginines at positions V:01 and V:05 with histidines [R208H; R212H]. The metal ion site conferred high-potency inverse agonist properties (EC(50), 1.7 microM) to Zn(II) in addition to agonist and allosteric enhancing properties at concentrations >10 microM. The chemokine interaction with [R208H;R212H]-ORF74 was altered compared with wild-type ORF74-HHV8 with decreased agonist (CXCL1/GROalpha) potency (84-fold), affinity (5.8- and 136-fold in competition against agonist and inverse agonist, respectively), and binding capacity (B(max); 25-fold). Zn(II) in activating concentrations (100 microM) acted as an allosteric enhancer as it increased the B(max) (7.1-fold), the potency (9.9-fold), the affinity (1.7- and 6.1-fold in competition against agonist and inverse agonist, respectively), and the efficacy (2.5-fold) of CXCL1/GROalpha. The activating properties of Zn(II) were not due to a metal ion site between the ligand and the receptor because CXCL1/GROalpha analogs in which the putative metal-ion binding residues had been substituted-[H19A] and [H34A]-acted like wild-type CXCL1/GROalpha. Based on the complex action of Zn(II) and on the chemokine interaction for [R208H;R212H]-ORF74, we conclude that the extracellular end of TM-V is important for the activation of this CXC-chemokine receptor.


Subject(s)
Protein Engineering , Receptors, Cell Surface/chemistry , Amino Acid Sequence , Base Sequence , Binding, Competitive , Circular Dichroism , Cloning, Molecular , DNA Primers , Enzyme-Linked Immunosorbent Assay , Escherichia coli/genetics , Humans , Molecular Sequence Data , Protein Conformation , Receptors, Cell Surface/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...