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.
Int J Oncol ; 36(6): 1401-8, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20428763

ABSTRACT

The aim of this study was to elucidate the role of CD97 isoforms in gastric carcinoma. Out of four gastric cancer cell lines investigated, BGC-823 cells demonstrating low CD97 protein expression were stably transfected with pcDNA3.1 vector containing CD97/EGF1,2,5 or CD97/EGF1,2,3,4,5 inserts. Behavior of transfected cells was systematically investigated by employing proliferation, motility and invasive assays. As a result, we found that over-expression of CD97/EGF1,2,5 isoform correlated with increased motile and invasive ability of the clones. Furthermore, CD97/EGF1,2,5 isoform over-expression (3.8 times higher) was followed by significant decrease of CD97/EGF1,2,3,4,5 isoform (10.3 times lower). In contrast, CD97/EGF1,2,3,4,5 clones revealed significantly reduced invasive properties as compared with corresponding controls. The changes in acetylation status were one of the possible mechanisms affecting behavior of transfected cells. We concluded from the study that CD97 is closely related with advanced stages and higher invasiveness of gastric carcinoma. The study further lightened the tumor promoting role of CD97 small isoform in cancer progression and indicated the possible suppressive properties of the full length isoform of CD97.


Subject(s)
Antigens, CD/genetics , Antigens, CD/metabolism , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Neoplasm Invasiveness/genetics , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Blotting, Western , Cell Line, Tumor , Cell Movement/genetics , Cell Separation , Flow Cytometry , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Neoplasm Invasiveness/pathology , Protein Isoforms , Receptors, G-Protein-Coupled , Reverse Transcriptase Polymerase Chain Reaction , Stomach Neoplasms/pathology , Transfection
2.
Mol Cancer Res ; 8(4): 494-506, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20332215

ABSTRACT

Relaxin increases cell motility and in vitro invasiveness in human thyroid carcinoma cells but the underlying molecular mechanisms of this action are largely unknown. In the present study, we show that relaxin transcriptionally upregulates the calcium-binding protein S100A4 (metastasin) and increases the cytosolic 10-kDa monomer and the 20-kDa dimer form of S100A4 in human thyroid carcinoma cells. The relaxin-induced increase in cell motility was blocked completely when S100A4 expression was diminished using an S100A4 small interfering RNA knockdown approach. We have shown previously the expression of the insulin-like family member relaxin in human thyroid carcinoma tissues but not in benign thyroid tissues. Human thyroid carcinoma tissues expressing relaxin also stained positive for S100A4. In nude mouse experiments, human thyroid carcinoma cell transfectants with constitutive expression of relaxin generated large and fast-growing tumors with significantly increased numbers of proliferating cells. We provide evidence in our cell model that the relaxin target protein S100A4 secreted by the thyroid carcinoma transfectants may not only enhance tumor cell motility but also promote xenograft angiogenesis as determined by the higher density of tumor microvessels and the angiogenic potential of S100A4 in in vitro tube formation assays. In conclusion, we have identified S100A4 as a major mediator of the actions of relaxin in thyroid carcinoma cell motility and in vivo thyroid tumor angiogenesis.


Subject(s)
Carcinoma/metabolism , Neoplasm Invasiveness/physiopathology , Relaxin/metabolism , S100 Proteins/genetics , Thyroid Neoplasms/metabolism , Animals , Carcinoma/chemically induced , Carcinoma/physiopathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/physiology , Cell Proliferation/drug effects , Cells, Cultured , Graft Survival/physiology , Humans , Male , Mice , Mice, Nude , Microcirculation/drug effects , Microcirculation/physiology , Neoplasm Transplantation , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/physiopathology , RNA Interference , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Relaxin/pharmacology , S100 Calcium-Binding Protein A4 , Thyroid Neoplasms/chemically induced , Thyroid Neoplasms/physiopathology , Transplantation, Heterologous , Up-Regulation/drug effects , Up-Regulation/genetics
3.
Int J Cancer ; 127(3): 521-31, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-19950223

ABSTRACT

The functional role of INSL3 and its receptor RXFP2 in carcinogenesis is largely unknown. We have previously demonstrated (pro-)cathepsin-L as a target of INSL3 in human thyroid cancer cells facilitating penetration of tumor cells through elastin matrices. We demonstrate the expression of RXFP2 in human thyroid tissues and in mouse follicular thyroid epithelial cells using Cre-recombinase transgene driven by Rxfp2 promoter. Recombinant and secreted INSL3 increased the motility of thyroid carcinoma (TC) cells in an autocrine/paracrine manner. This effect required the presence of RXFP2. We identified S100A4 as a novel INSL3 target molecule and showed that S100A4 facilitated INSL3-induced enhanced motility. Stable transfectants of the human follicular TC cell line FTC-133 expressing and secreting bioactive human INSL3 displayed enhanced anchorage-independent growth in soft agar assays. Xenotransplant experiments in nude mice showed that INSL3, but not EGFP-mock transfectants, developed fast-growing and highly vascularized xenografts. We used human umbilical vein endothelial cells in capillary tube formation assays to demonstrate increased 2-dimensional tube formations induced by recombinant human INSL3 and human S100A4 comparable to the effect of vascular endothelial growth factor used as positive control. We conclude that INSL3 is a powerful and multifunctional promoter of tumor growth and angiogenesis in human thyroid cancer cell xenografts. INSL3 actions involve RXFP2 activation and the secretion of S100A4 and (pro-)cathepsin-L.


Subject(s)
Cell Transformation, Neoplastic , Insulin/physiology , Proteins/physiology , Thyroid Neoplasms/physiopathology , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Base Sequence , Cell Division/physiology , Child , DNA Primers , Female , Humans , In Situ Nick-End Labeling , Male , Mice , Mice, Nude , Middle Aged , Neoplasm Metastasis , RNA, Small Interfering , S100 Calcium-Binding Protein A4 , S100 Proteins/metabolism , Thyroid Neoplasms/pathology , Transplantation, Heterologous , Young Adult
4.
Breast Cancer Res ; 10(4): R71, 2008.
Article in English | MEDLINE | ID: mdl-18718015

ABSTRACT

INTRODUCTION: Relaxin levels are increased in cases of human breast cancer and has been shown to promote cancer cell migration in carcinoma cells of the breast, prostate gland and thyroid gland. In oestrogen receptor alpha-negative MDA-MB-231 human breast cancer cells, relaxin was shown to down-regulate the metastasis-promoting protein S100A4 (metastasin), a highly significant prognostic factor for poor survival in breast cancer patients. The cellular mechanisms of relaxin exposure in breast cancer cells are not fully understood. The aim of this study was to investigate short-term and long-term effects of relaxin on cancer cell motility and S100A4 expression and to determine the long-term effects of relaxin on in vivo tumour growth in an oestrogen-independent context. METHOD: We have established stable transfectants of highly invasive oestrogen-receptor alpha-negative MDA-MB-231 human breast cancer cells with constitutive expression of bioactive H2-relaxin (MDA/RLN2). RLN2 secretion was determined by ELISA. Relaxin receptor RXFP1 (Relaxin-family-peptide) was detected by reverse transcription (RT) PCR and its activation was assessed by induction of cyclic adenosine monophosphate (cAMP). Stable MDA/RLN2 clones and RLN2 treated MDA-MB-231 cells were subjected to motility and in vitro-invasion assays. Proliferation was assessed in bromodeoxyuridine (BrdU) and MTT assays. S100A4 expression was determined by RT-PCR and Western blot. Specific small interfering RNA was employed to down-regulate relaxin receptor and S100A4. MDA/EGFP vector control and two MDA/RLN2 clones were injected subcutaneously in nude mice to determine tumour growth and cancer cell invasiveness in vivo. Xenograft tumour tissues were assessed by histology and immunohistochemistry and frozen tissues were used for the detection of S100A4 and RLN2. RESULTS: Short-term exposure to relaxin for 24 hours increased cell motility in a relaxin receptor-dependent manner. This increase in cell motility was mediated by S100A4. Long-term exposure to relaxin secreted from stable transfectants reduced cell motility and in vitro invasiveness. Relaxin decreased cell proliferation and down-regulated cellular S100A4 levels in MDA-MB-231 and T47D breast cancer cells. Stable MDA/RLN2 transfectants produced smaller xenograft tumours containing reduced S100A4 protein levels in vivo. CONCLUSION: Our results indicate that long-term exposure to relaxin confers growth inhibitory and anti-invasive properties in oestrogen-independent tumours in vivo, which may in part be mediated through a down-regulation of S100A4.


Subject(s)
Breast Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Relaxin/physiology , Animals , Breast Neoplasms/pathology , Carcinoma/metabolism , Carcinoma/pathology , Cell Line, Tumor , Humans , Male , Mice , Mice, Nude , Neoplasm Metastasis , Neoplasm Transplantation , Prognosis , Receptors, Estrogen/metabolism , Relaxin/metabolism , S100 Calcium-Binding Protein A4 , S100 Proteins/metabolism
5.
Adv Exp Med Biol ; 612: 104-18, 2007.
Article in English | MEDLINE | ID: mdl-18161484

ABSTRACT

Relaxin and INSL3 are novel autocrine/paracrine insulin-like hormones in tumor biology. Both effectors can bind to and activate the leucine-rich G-protein coupled receptors LGR7 relaxin receptor) or LGR8 (relaxin/INSL3 receptor). These relaxin-like ligand-receptor systems modulate cellular functions and activate signaling cascades in a tumor-specific context leading to changes in tumor cell proliferation, altered motility/migration and enhanced production/secretion ofpotent proteolytic enzymes. Matrix-metalloproteinases (MMP), tissue inhibitors of metalloproteinases (TIMP) and acid hydrolases such as cathepsins can facilitate tissue degradation and represent important proteolytic mediators of relaxin-like actions on tumor cell invasion and metastasis. This review presents recent new findings and emphasises the important functions of the relaxin/INSL3 ligand-receptor system as novel autocrine/paracrine effectors influencing tumor progression and tissue invasiveness.


Subject(s)
Autocrine Communication , Insulin/metabolism , Neoplasm Proteins/metabolism , Neoplasms/metabolism , Paracrine Communication , Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Cell Movement , Disease Progression , Female , Humans , Ligands , Male , Neoplasm Invasiveness , Neoplasms/pathology , Signal Transduction
6.
Int J Oncol ; 27(2): 307-15, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16010410

ABSTRACT

The human prostate gland is a well known source of H1 and H2 relaxin but information is lacking on the expression and potential role of the INSL3 peptide hormone within the prostate gland. In the present study we have investigated the expression of human INSL3 in patients with benign prostate hyperplasia (BPH), prostate intraepithelial neoplasia (PIN) and prostate carcinoma tissues. Of the prostate epithelial cells, strongest INSL3 expression was detected in the basal epithelial cell compartment. Weaker INSL3 mRNA expression and immunoreactive INSL3 production were observed in secretory epithelial cells and in interstitial smooth muscle cells. Prostate epithelial cells were also a source for transcripts encoding the INSL3 receptor LGR8 suggesting the presence of an autocrine/paracrine INSL3-LGR8 ligand-receptor system within the human prostate. Three human prostate carcinoma cell lines displayed differential gene activity for INSL3 and LGR8. While LNCaP was devoid of INSL3, the androgen-insensitive PC-3 and the stromal prostate cell line hPCP co-expressed INSL3 and LGR8 transcripts. In addition to expressing INSL3 mRNA, the LGR8-negative DU-145 also expressed an INSL3 splice form formerly demonstrated in thyroid carcinoma cells. When incubated with recombinant INSL3, PC-3 cells showed significantly increased intracellular cAMP levels indicating functional LGR8 receptors. INSL3 did not alter the proliferative or metabolic activity of PC-3 carcinoma cells. Instead, PC-3 responded to INSL3 with significantly enhanced tumor cell motility and a transcriptional down-regulation of ErbB receptors and EGF. All-trans-retinoic acid was demonstrated in PC-3 to up-regulate LGR8 gene activity in a dose- and time-dependent manner while having no effect on INSL3 gene activity. In conclusion, we have identified a functional INSL3-LGR8 ligand-receptor system in human prostate carcinoma cells.


Subject(s)
Insulin/genetics , Prostatic Hyperplasia/pathology , Prostatic Intraepithelial Neoplasia/pathology , Prostatic Neoplasms/pathology , Proteins/genetics , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cyclic AMP/metabolism , Dose-Response Relationship, Drug , Epidermal Growth Factor/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Gene Expression/drug effects , Humans , Immunohistochemistry , In Situ Hybridization , Insulin/analysis , Insulin/pharmacology , Male , Prostatic Hyperplasia/genetics , Prostatic Hyperplasia/metabolism , Prostatic Intraepithelial Neoplasia/genetics , Prostatic Intraepithelial Neoplasia/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Proteins/analysis , Proteins/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, G-Protein-Coupled/analysis , Receptors, G-Protein-Coupled/genetics , Recombinant Proteins/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Tretinoin/pharmacology
7.
Ann N Y Acad Sci ; 1041: 449-61, 2005 May.
Article in English | MEDLINE | ID: mdl-15956746

ABSTRACT

We investigated the expression of H1, H2 relaxin and INSL-3, mRNA and protein, and LGR7 and LGR8 transcripts in human C-cell hyperplasia, primary medullary thyroid carcinoma (MTC) tissues, MTC metastases, and the human MTC-TT and mouse MTC-M cell lines. Relaxin-like peptide hormones were detected in C-cell hyperplasia and in MTC tissues, but were absent in human normal parafollicular C-cells of benign goiter tissues. In contrast to calcitonin, mRNA, and immunoreactive protein, no differences in the expression of relaxin and INSL3 were observed in MTC tissues of different pTNM classification or between primary and metastatic MTC tissues studied. All MTC tissues constitutively expressed LGR7 and LGR8 transcripts. Thus, relaxin-like hormones appear to be present early during C-cell hyperplasia and potentially functional relaxin/INSL3 ligand-receptor systems are present in human MTC tissues and in MTC cell lines.


Subject(s)
Carcinoma, Medullary/metabolism , Relaxin/metabolism , Carcinoma, Medullary/genetics , Carcinoma, Medullary/pathology , Cell Line, Tumor , Humans , In Situ Hybridization , Lymphatic Metastasis , Relaxin/genetics , Transcription, Genetic/genetics
8.
Ann N Y Acad Sci ; 1041: 462-9, 2005 May.
Article in English | MEDLINE | ID: mdl-15956747

ABSTRACT

Expressed in the human breast and in human breast cancer tissues, the heterodimeric peptide hormone relaxin is involved in extracellular matrix turnover. To investigate the role of relaxin in estrogen receptor-alpha negative human breast cancer cells, we established transfectants of the human MDA-MB-231 breast cancer cell line stably overexpressing H2-relaxin (MDA-MB-231/pIRES-EGFP-H2). These transfectants produced and secreted functional relaxin. Our investigations revealed downregulation of mRNA and protein of the calcium binding protein S100A4 (metastasin) in MDA-MB-231/pIRES-EGFP-H2 transfectants, indicating a distinct and novel role for relaxin in estrogen receptor-alpha negative human breast cancer cells.


Subject(s)
Breast Neoplasms/metabolism , Down-Regulation , Relaxin/metabolism , S100 Proteins/metabolism , Breast Neoplasms/genetics , Cell Line, Tumor , Humans , Relaxin/genetics , S100 Calcium-Binding Protein A4 , S100 Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...