Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
Int J Mol Sci ; 21(14)2020 Jul 21.
Article in English | MEDLINE | ID: mdl-32708166

ABSTRACT

During the metastasis process, tumor cells invade the blood circulatory system directly from venous capillaries or indirectly via lymphatic vessels. Understanding the relative contribution of each pathway and identifying the molecular targets that affect both processes is critical for reducing cancer spread. Methionine aminopeptidase 2 (MetAp2) is an intracellular enzyme known to modulate angiogenesis. In this study, we investigated the additional role of MetAp2 in lymphangiogenesis. A histological staining of tumors from human breast-cancer donors was performed in order to detect the level and the localization of MetAp2 and lymphatic capillaries. The basal enzymatic level and activity in vascular and lymphatic endothelial cells were compared, followed by loss of function studies determining the role of MetAp2 in lymphangiogenesis in vitro and in vivo. The results from the histological analyses of the tumor tissues revealed a high MetAp2 expression, with detectable sites of co-localization with lymphatic capillaries. We showed slightly reduced levels of the MetAp2 enzyme and MetAp2 mRNA expression and activity in primary lymphatic cells when compared to the vascular endothelial cells. The genetic and biochemical manipulation of MetAp2 confirmed the dual activity of the enzyme in both vascular and lymphatic remodulation in cell function assays and in a zebrafish model. We found that cancer-related lymphangiogenesis is inhibited in murine models following MetAp2 inhibition treatment. Taken together, our study provides an indication that MetAp2 is a significant contributor to lymphangiogenesis and carries a dual role in both vascular and lymphatic capillary formation. Our data suggests that MetAp2 inhibitors can be effectively used as anti-metastatic broad-spectrum drugs.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Cell Proliferation/genetics , Endothelial Cells/metabolism , Lymphangiogenesis/genetics , Lymphatic Metastasis/genetics , Methionyl Aminopeptidases/metabolism , Neovascularization, Pathologic/metabolism , Animals , Animals, Genetically Modified , Cell Line, Tumor , Cell Proliferation/drug effects , Endothelial Cells/drug effects , Endothelial Cells/enzymology , Endothelium, Vascular/drug effects , Endothelium, Vascular/enzymology , Endothelium, Vascular/metabolism , Humans , Lymphatic Metastasis/pathology , Lymphatic Vessels/drug effects , Lymphatic Vessels/metabolism , Male , Methionyl Aminopeptidases/antagonists & inhibitors , Methionyl Aminopeptidases/genetics , Mice , Mice, Inbred C57BL , Neovascularization, Pathologic/enzymology , O-(Chloroacetylcarbamoyl)fumagillol/pharmacology , Xenograft Model Antitumor Assays , Zebrafish
2.
Cell Mol Life Sci ; 71(13): 2517-33, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24177339

ABSTRACT

Mammalian protease-activated-receptor-1 and -2 (PAR1 and PAR2) are activated by proteases found in the flexible microenvironment of a tumor and play a central role in breast cancer. We propose in the present study that PAR1 and PAR2 act together as a functional unit during malignant and physiological invasion processes. This notion is supported by assessing pro-tumor functions in the presence of short hairpin; shRNA knocked-down hPar2 or by the use of a truncated PAR2 devoid of the entire cytoplasmic tail. Silencing of hPar2 by shRNA-attenuated thrombin induced PAR1 signaling as recapitulated by inhibiting the assembly of Etk/Bmx or Akt onto PAR1-C-tail, by thrombin-instigated colony formation and invasion. Strikingly, shRNA-hPar2 also inhibited the TFLLRN selective PAR1 pro-tumor functions. In addition, while evaluating the physiological invasion process of placenta extravillous trophoblast (EVT) organ culture, we observed inhibition of both thrombin or the selective PAR1 ligand; TFLLRNPNDK induced EVT invasion by shRNA-hPar2 but not by scrambled shRNA-hPar2. In parallel, when a truncated PAR2 was utilized in a xenograft mouse model, it inhibited PAR1-PAR2-driven tumor growth in vivo. Similarly, it also attenuated the interaction of Etk/Bmx with the PAR1-C-tail in vitro and decreased markedly selective PAR1-induced Matrigel invasion. Confocal images demonstrated co-localization of PAR1 and PAR2 in HEK293T cells over-expressing YFP-hPar2 and HA-hPar1. Co-immuno-precipitation analyses revealed PAR1-PAR2 complex formation but no PAR1-CXCR4 complex was formed. Taken together, our observations show that PAR1 and PAR2 act as a functional unit in tumor development and placenta-uterus interactions. This conclusion may have significant consequences on future breast cancer therapeutic modalities and improved late pregnancy outcome.


Subject(s)
Breast Neoplasms/genetics , Receptor, PAR-1/genetics , Receptor, PAR-2/genetics , Tumor Microenvironment/genetics , Animals , Breast Neoplasms/pathology , Breast Neoplasms/therapy , Female , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Mice , Oligopeptides/metabolism , Pregnancy , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptor, PAR-1/metabolism , Receptor, PAR-2/metabolism , Receptors, CXCR4/genetics , Xenograft Model Antitumor Assays
3.
FASEB J ; 26(5): 2031-42, 2012 May.
Article in English | MEDLINE | ID: mdl-22291441

ABSTRACT

A pivotal role is attributed to the estrogen-receptor (ER) pathway in mediating the effect of estrogen in breast cancer progression. Yet the precise mechanisms of cancer development by estrogen remain poorly understood. Advancing tumor categorization a step forward, and identifying cellular gene fingerprints to accompany histopathological assessment may provide targets for therapy as well as vehicles for evaluating the response to treatment. We report here that in breast carcinoma, estrogen may induce tumor development by eliciting protease-activated receptor-1 (PAR(1)) gene expression. Induction of PAR(1) was shown by electrophoretic mobility shift assay, luciferase reporter gene driven by the hPar(1) promoter, and chromatin-immunoprecipitation analyses. Functional estrogen regulation of hPar1 in breast cancer was demonstrated by an endothelial tube-forming network. Notably, tissue-microarray analyses from an established cohort of women diagnosed with invasive breast carcinoma exhibited a significantly shorter disease-free (P=0.006) and overall (P=0.02) survival of patients that were positive for ER and PAR(1), compared to ER-positive but PAR(1)-negative patients. We propose that estrogen transcriptionally regulates hPar(1), culminating in an aggressive gene imprint in breast cancer. While ER(+) patients are traditionally treated with hormone therapy, the presence of PAR(1) identifies a group of patients that requires additional treatment, such as anti-PAR(1) biological vehicles or chemotherapy.


Subject(s)
Breast Neoplasms/metabolism , Estrogens/physiology , Gene Expression Regulation/physiology , Receptor, PAR-1/genetics , Base Sequence , Breast Neoplasms/pathology , Chromatin Immunoprecipitation , Cohort Studies , DNA , DNA Primers , Electrophoretic Mobility Shift Assay , Enzyme-Linked Immunosorbent Assay , Female , Humans , Molecular Sequence Data , Polymerase Chain Reaction , Promoter Regions, Genetic
5.
Eur J Obstet Gynecol Reprod Biol ; 185: 13-8, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25496845

ABSTRACT

BACKGROUND: Recurrent pregnancy loss (RPL) has been associated with impaired maternal-fetal communication. Protease-activated-receptor 1 (PAR1) is critical for trophoblast invasion and establishment unrelated to its role in vascular biology. OBJECTIVES: To analyze whether polymorphisms of PAR1 [-1426C/T], [-506I/D], and/or IVS[-14A/T] are associated with unexplained RPL. PATIENTS/METHODS: A case-control pilot study conducted in 39 healthy women with history of unexplained RPL and 98 women with a full-term, uncomplicated deliveries and no history of RPL. RESULTS: Women with RPL were significantly more likely to be heterozygous for [-1426C/T] (12.8% versus 3.2%; p=0.049); the heterozygous state for IVS[-14A/T] was also more common (15.4% versus 4.4%; p=0.064). There was no difference between groups for [-506I/D] genotypes. The functional consequence for [-1426C/T] and IVS[-14A/T] polymorphisms is underscored by the markedly low PAR1 mRNA levels in those women. Bioinformatics indicate generation of a new consensus motif for repressor Kruppel-like factor 3 (KLF3) in [-1426T]. Moreover, chromatin immunoprecipitation (ChIP) analysis confirmed a physical association between KLF3 protein and the hPar1 DNA obtained from women with the [-1426C/T] polymorphism. CONCLUSIONS: We hypothesize that the significantly low PAR1 levels impact placenta establishment and consequently pregnancy outcome, thereby profiling a novel risk factor for unexplained RPL.


Subject(s)
Abortion, Habitual/genetics , Receptor, PAR-1/genetics , Abortion, Habitual/metabolism , Adult , Base Sequence , Case-Control Studies , Female , Humans , Kruppel-Like Transcription Factors/metabolism , Molecular Sequence Data , Pilot Projects , Polymorphism, Genetic , Pregnancy
6.
Immunology ; 105(1): 35-46, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11849313

ABSTRACT

Recent characterization of the thrombin receptor indicates that it plays a role in T-cell signalling pathways. However, little is known regarding the signalling events following stimulation of additional members of the protease-activated receptor (PAR) family, i.e. PAR2 and PAR3. Most of the postligand cascades are largely unknown. Here, we illustrate that in Jurkat T-leukaemic cells, activation of PAR1, PAR2 and PAR3 induce tyrosine phosphorylation of Vav1. This response was impaired in Jurkat T cells deficient in p56lck (JCaM1.6). Activation of PARs also led to an increase in tyrosine phosphorylation of ZAP-70 and SLP-76, two key proteins in T-cell receptor (TCR) signalling. We also demonstrated that p56lck is meaningful for integrin signalling. Thus, JCaM1.6 cells exhibited a marked reduction in their adherence to fibronectin-coated plates, as compared to the level of adherence of Jurkat T cells. While the phosphorylation of Vav1 in T cells is augmented following adhesion, no additional increase was noted following treatment of the adhered cells with PARs. Altogether, we have identified key components in the postligand-signalling cascade of PARs and integrins. Furthermore, we have identified Lck as a critical and possibly upstream component of PAR-induced Vav1 phosphorylation, as well as integrin activation, in Jurkat T cells.


Subject(s)
Integrins/physiology , Receptors, Thrombin/physiology , Signal Transduction/physiology , T-Lymphocytes/physiology , Autoradiography , Cell Adhesion/physiology , Cells, Cultured , Electrophoresis, Polyacrylamide Gel , Humans , Luminescent Measurements , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/physiology , Phosphorylation , Receptor, PAR-1 , Receptor, PAR-2 , Tyrosine/physiology , Vasopressins/physiology
7.
Gynecol Oncol ; 84(2): 296-302, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11812090

ABSTRACT

OBJECTIVE: The goal of this work was to evaluate the involvement of gonadotropins in the regulation of adhesion of human epithelial ovarian carcinoma. We studied two pathways that were previously implicated in the metastatic implantation of ovarian carcinoma to the peritoneum, namely hyaluronan-CD44 and RGD-integrin mediated adhesion. METHODS: Two cell lines derived from human epithelial ovarian carcinoma (MLS and OC238) were stimulated with luteinizing hormone (LH) and/or follicle stimulating hormone (FSH). Expression of CD44 was evaluated by Western blotting. Expression of alpha(v)-integrins was studied by RT-PCR and Northern blot. Integrin and CD44 mediated adhesion of the cells was analyzed using culture plates coated either with a thrombin derived RGD containing peptide or fibronectin for integrin mediated adhesion or with hyaluronan for CD44 mediated adhesion. RESULTS: MLS cells stimulated with either LH or FSH showed increased adhesion to culture plates coated with hyaluronan, as well as to culture plates coated with fibronectin or with a thrombin derived RGD containing peptide. In these cells, gonadotropin stimulation led to induced expression of the integrin subunit alpha(v) and CD44, the cell surface hyaluronan receptor. On the other hand, OC238 cells showed no expression of the integrin subunit alpha(v) and no hormonal effect on the expression of CD44. Accordingly, adhesion of OC238 cells on either RGD or CD44 was not affected by hormonal stimulation. CONCLUSION: Elevated levels of gonadotropins may in some cases facilitate peritoneal metastatic dissemination of ovarian cancer by increasing cell adhesion, the first essential step in the invasion process.


Subject(s)
Antigens, CD/physiology , Follicle Stimulating Hormone/pharmacology , Hyaluronan Receptors/physiology , Luteinizing Hormone/pharmacology , Ovarian Neoplasms/pathology , Antigens, CD/biosynthesis , Blotting, Western , Cell Adhesion/drug effects , Cell Adhesion/physiology , Epithelial Cells/pathology , Female , Follicle Stimulating Hormone/metabolism , Follicle Stimulating Hormone/physiology , Humans , Hyaluronan Receptors/biosynthesis , Hyaluronic Acid/pharmacology , Integrin alphaV , Luteinizing Hormone/metabolism , Luteinizing Hormone/physiology , Oligopeptides/pharmacology , Ovarian Neoplasms/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
8.
J Pathol ; 200(1): 47-52, 2003 May.
Article in English | MEDLINE | ID: mdl-12692840

ABSTRACT

Human fetal development depends on the ability of the embryo to gain access to the maternal circulation. Thus, specialized stem cells of the newly formed placenta, trophoblast, invade the uterus and its arterial network to establish an efficient feto-maternal molecular exchange. To accomplish this task, trophoblast differentiation during the first trimester of pregnancy involves cell proliferation, invasion, and extracellular matrix (ECM) remodelling. Trophoblast invasion shares many features with tumour cell invasion, with the distinction that it is strictly spatially and temporally controlled. We have previously demonstrated that PAR1, the first member of the protease-activated receptor (PAR) family, plays a central role in tumour cell invasion. In the present study we have examined the pattern of expression of PAR1 and other PAR family candidates during early human placental development. We show that PAR1 and PAR3 are highly and spatially expressed between the 7th and 10th weeks of gestation but not at the 12th week and thereafter. Likewise, high expression levels of PAR1 and PAR3 were observed in the cytotrophoblast cells of complete hydatidiform mole as compared to minimal levels in normal age-matched placenta. Together, our data suggest the involvement of PAR1 and PAR3 in restricted and unrestricted pathological trophoblast invasion.


Subject(s)
Placenta/metabolism , Receptors, Thrombin/analysis , Trophoblasts/physiology , Chorionic Villi/metabolism , Down-Regulation/physiology , Extracellular Matrix/metabolism , Female , Humans , Hydatidiform Mole/metabolism , Immunohistochemistry/methods , In Situ Hybridization/methods , Maternal-Fetal Exchange/physiology , Polymerase Chain Reaction/methods , Pregnancy , Pregnancy Proteins/analysis , Pregnancy Trimester, First/physiology , Receptor, PAR-1 , Trophoblasts/cytology
SELECTION OF CITATIONS
SEARCH DETAIL