Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
J Am Chem Soc ; 137(35): 11365-75, 2015 Sep 09.
Article in English | MEDLINE | ID: mdl-26317395

ABSTRACT

Peptides can be developed as effective antagonists of protein-protein interactions, but conventional peptides (i.e., oligomers of l-α-amino acids) suffer from significant limitations in vivo. Short half-lives due to rapid proteolytic degradation and an inability to cross cell membranes often preclude biological applications of peptides. Oligomers that contain both α- and ß-amino acid residues ("α/ß-peptides") manifest decreased susceptibility to proteolytic degradation, and when properly designed these unnatural oligomers can mimic the protein-recognition properties of analogous "α-peptides". This report documents an extension of the α/ß-peptide approach to target intracellular protein-protein interactions. Specifically, we have generated α/ß-peptides based on a "stapled" Bim BH3 α-peptide, which contains a hydrocarbon cross-link to enhance α-helix stability. We show that a stapled α/ß-peptide can structurally and functionally mimic the parent stapled α-peptide in its ability to enter certain types of cells and block protein-protein interactions associated with apoptotic signaling. However, the α/ß-peptide is nearly 100-fold more resistant to proteolysis than is the parent stapled α-peptide. These results show that backbone modification, a strategy that has received relatively little attention in terms of peptide engineering for biomedical applications, can be combined with more commonly deployed peripheral modifications such as side chain cross-linking to produce synergistic benefits.


Subject(s)
Cell-Penetrating Peptides/chemistry , Cell-Penetrating Peptides/pharmacology , Intracellular Space/drug effects , Intracellular Space/metabolism , Protein Folding , Amino Acid Sequence , Animals , Apoptosis Regulatory Proteins/chemistry , Bcl-2-Like Protein 11 , Cell Membrane Permeability , Cell Survival/drug effects , Cell-Penetrating Peptides/metabolism , Cytochromes c/metabolism , HCT116 Cells , Humans , Membrane Proteins/chemistry , Mice , Models, Molecular , Molecular Sequence Data , Peptide Hydrolases/metabolism , Protein Binding/drug effects , Protein Stability , Protein Structure, Tertiary , Proteolysis , Proto-Oncogene Proteins/chemistry
2.
Int J Cancer ; 121(1): 6-11, 2007 Jul 01.
Article in English | MEDLINE | ID: mdl-17354225

ABSTRACT

The pro-inflammatory cytokine, tumour necrosis factor-alpha, TNF-alpha, is dysregulated in malignant compared with normal ovarian surface epithelium (OSE). Several epidemiological studies have associated inflammation with ovarian tumorigenesis, with TNF-alpha playing a key role in modulating invasion, angiogenesis and metastasis. Here, we show that TNF-alpha also induces expression of arate-limiting enzyme in arginine synthesis, argininosuccinate synthetase (AS), thereby linking inflammation with several arginine-dependent metabolic pathways, implicated in accelerated carcinogenesis and tumour progression. Having identified AS mRNA induction in TNF-alpha-treated IGROV-1 ovarian cancer cells, using RNA-arbitrarily primed-PCR, we then observed differential regulation of AS mRNA and protein in malignant, compared with normal, OSE cells. A cDNA cancer profiling array with matched normal ovarian and ovarian tumour samples revealed increased expression of AS mRNA in the latter. Moreover, AS protein co-localised with TNF-alpha in ovarian cancer cells, with significantly higher levels of AS in malignant compared with normal ovarian tissue. Increased co-expression of AS and TNF-alpha mRNA was also observed in 2 other epithelial tumours, non-small cell lung and stomach cancer, compared with normal corresponding tissues. In summary, high levels of AS expression, which may be required for several arginine-dependent processes in cancer, including the production of nitric oxide, proline, pyrimidines and polyamines, is regulated by TNF-alpha and may provide an important molecular pathway linking inflammation and metabolism to ovarian tumorigenesis.


Subject(s)
Argininosuccinate Synthase/metabolism , Epithelial Cells/drug effects , Epithelial Cells/enzymology , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Ovarian Neoplasms/enzymology , Tumor Necrosis Factor-alpha/pharmacology , Argininosuccinate Synthase/genetics , Epithelial Cells/cytology , Female , Health , Humans , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , RNA, Messenger/genetics , Tumor Cells, Cultured
3.
Cancer Res ; 67(2): 585-92, 2007 Jan 15.
Article in English | MEDLINE | ID: mdl-17234767

ABSTRACT

Constitutive expression of the inflammatory cytokine tumor necrosis factor-alpha (TNF-alpha) is characteristic of malignant ovarian surface epithelium. We investigated the hypothesis that this autocrine action of TNF-alpha generates and sustains a network of other mediators that promote peritoneal cancer growth and spread. When compared with two ovarian cancer cell lines that did not make TNF-alpha, constitutive production of TNF-alpha was associated with greater release of the chemokines CCL2 and CXCL12, the cytokines interleukin-6 (IL-6) and macrophage migration-inhibitory factor (MIF), and the angiogenic factor vascular endothelial growth factor (VEGF). TNF-alpha production was associated also with increased peritoneal dissemination when the ovarian cancer cells were xenografted. We next used RNA interference to generate stable knockdown of TNF-alpha in ovarian cancer cells. Production of CCL2, CXCL12, VEGF, IL-6, and MIF was decreased significantly in these cells compared with wild-type or mock-transfected cells, but in vitro growth rates were unaltered. Tumor growth and dissemination in vivo were significantly reduced when stable knockdown of TNF-alpha was achieved. Tumors derived from TNF-alpha knockdown cells were noninvasive and well circumscribed and showed high levels of apoptosis, even in the smallest deposits. This was reflected in reduced vascularization of TNF-alpha knockdown tumors. Furthermore, culture supernatants from such cells failed to stimulate endothelial cell growth in vitro. We conclude that autocrine production of TNF-alpha by ovarian cancer cells stimulates a constitutive network of other cytokines, angiogenic factors, and chemokines that may act in an autocrine/paracrine manner to promote colonization of the peritoneum and neovascularization of developing tumor deposits.


Subject(s)
Ovarian Neoplasms/pathology , Tumor Necrosis Factor-alpha/physiology , Animals , Cell Growth Processes/physiology , Cell Line, Tumor , Chemokine CXCL12 , Chemokines, CXC/metabolism , Female , Humans , Mice , Mice, Nude , Neoplasm Invasiveness , Neovascularization, Pathologic/immunology , Neovascularization, Pathologic/metabolism , Ovarian Neoplasms/blood supply , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Receptors, CXCR4/metabolism , Transfection , Transplantation, Heterologous , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
4.
Cancer Res ; 66(24): 11802-7, 2006 Dec 15.
Article in English | MEDLINE | ID: mdl-17178876

ABSTRACT

Endothelin expression is increased in breast tumors and is associated with invasion and metastasis, whereas CCR7 expression by breast tumor cells may have a role in the organ specificity of breast cancer spread. In this article, we have analyzed whether endothelins influence breast tumor cell expression of the chemokine receptor CCR7. Stimulation of human breast tumor cell lines with endothelins increased cell surface expression of CCR7 via endothelin receptor A. The iron chelators desferrioxamine and cobalt chloride, which induce hypoxia-inducible factor (HIF)-mediated transcription, also increased CCR7 expression; transfection of a dominant-negative version of the HIF regulatory subunit, HIF-1alpha, into MCF-7 cells abolished CCR7 induction by endothelins, indicating that increased expression is due to HIF-1 stabilization. Endothelin stimulation promoted invasion toward the CCR7 ligands CCL19 and CCL21. Endothelin-mediated chemokine-independent invasion itself is dependent on CCR7 activity and could be abolished using a CCR7-neutralizing monoclonal antibody. In human breast carcinomas, mRNA expression of endothelins correlated with the level of CCR7 expression, both of which were associated with the presence of lymph node metastases. Expression of the CCR7 ligands CCL19 and CCL21 was also higher in breast cancer patients with lymph node involvement compared with those without, but expression of these chemokines did not correlate with endothelin expression. These data show that CCR7 may be regulated by the breast tumor microenvironment and further support the use of endothelin receptor antagonists in the treatment of invasive and metastatic breast cancer.


Subject(s)
Breast Neoplasms/genetics , Endothelin-1/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Receptors, Chemokine/genetics , Breast Neoplasms/pathology , Breast Neoplasms/surgery , Cell Line, Tumor , Cell Movement , DNA Primers , Female , Gene Expression Regulation, Neoplastic , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Neoplasm Invasiveness , Polymerase Chain Reaction , RNA, Messenger/genetics , RNA, Neoplasm/genetics , Receptors, CCR7
5.
Adv Drug Deliv Rev ; 58(8): 962-74, 2006 Oct 01.
Article in English | MEDLINE | ID: mdl-16996642

ABSTRACT

Chemokine gradients are central to the movement of cells in both homeostatic and pathological processes. Most cancers express a complex array of chemokines that influence the local microenvironment through recruitment of stromal cells and by stimulating angiogenesis. Recently, the discovery of chemokine receptors on tumor cells has led to speculation that the chemokine system may be involved in cancer cell growth and survival, and possibly the development of site-specific spread. Understanding the networks of chemokines and their receptors in cancer will enable manipulation of this system. Both chemokines and their receptors represent targets for therapeutic intervention either with antibodies or small molecule antagonists. However, due to the complexity of the system, and the number of chemokines and receptors that are also expressed by normal cells, issues remain concerning whether systemic or local drug delivery are preferable and whether the redundancy of the system will compensate if one chemokine or receptor is targeted. Nevertheless, efficacy has been demonstrated in a number of experimental models. By targeting this network, it may be possible to generate anti-tumor immune responses by altering the chemokine and/or leukocyte balance in tumors; alternatively, chemokine/chemokine receptor-expressing cancer cells could be directly targeted.


Subject(s)
Chemokines/immunology , Neoplasms/immunology , Receptors, Chemokine/immunology , Animals , Humans , Immunotherapy , Neoplasms/therapy
6.
Mol Cancer Ther ; 5(2): 382-90, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16505113

ABSTRACT

Epidemiologic studies implicate inflammatory stimuli in the development of ovarian cancer. The proinflammatory cytokine tumor necrosis factor alpha (TNF-alpha) and both its receptors (TNFRI and TNFRII) are expressed in biopsies of this malignancy. Here, we tested the hypothesis that TNF-alpha is a regulator of the proinflammatory microenvironment of ovarian cancer. A cancer profiling array showed higher expression of TNF-alpha in ovarian tumors compared with normal ovarian tissue, and cultured ovarian cancer cells expressed up to 1,000 times more TNF-alpha mRNA than cultured normal ovarian surface epithelial cells; TNF-alpha protein was only detected in the supernatant of tumor cell cultures. Treatment with TNF-alpha induced TNF-alpha mRNA via TNFRI in both malignant and normal cells with evidence for enhanced TNF-alpha mRNA stability in tumor cells. TNF-alpha induced TNF-alpha protein in an autocrine fashion in tumor but not in normal ovarian surface epithelial cells. The TNF-alpha neutralizing antibody infliximab reduced the constitutive levels of TNF-alpha mRNA in tumor cell lines capable of autocrine TNF-alpha production. Apart from TNF-alpha mRNA expression, several other proinflammatory cytokines were constitutively expressed in malignant and normal ovarian surface epithelial cells, including interleukin (IL)-1alpha, IL-6, CCL2, CXCL8, and M-CSF. TNF-alpha treatment further induced these cytokines with de novo transcription of IL-6 mRNA contrasting with the increased stability of CCL2 mRNA. RNA interference directed against TNF-alpha was highly effective in abolishing constitutive IL-6 production by ovarian tumor cells. In summary, we show that TNF-alpha is differentially regulated in ovarian cancer cells compared with untransformed cells and modulates production of several cytokines that may promote ovarian tumorigenesis. Infliximab treatment may have a role in suppressing the TNF-alpha-driven inflammatory response associated with ovarian cancer.


Subject(s)
Cytokines/metabolism , Ovarian Neoplasms/metabolism , Ovary/metabolism , Tumor Necrosis Factor-alpha/metabolism , Antibodies, Monoclonal/pharmacology , Cell Line, Tumor , Chemokine CCL2/metabolism , Cytokines/genetics , Epithelium/drug effects , Epithelium/metabolism , Female , Humans , Infliximab , Interleukin-6/metabolism , Ovarian Neoplasms/genetics , Ovary/drug effects , RNA Interference , RNA Stability , RNA, Messenger/analysis , RNA, Messenger/metabolism , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/genetics
7.
Cancer Res ; 65(22): 10355-62, 2005 Nov 15.
Article in English | MEDLINE | ID: mdl-16288025

ABSTRACT

Epithelial ovarian cancer cells express the chemokine receptor, CXCR4, which may be associated with increased survival and metastatic potential, but the regulation of this receptor is not understood. The inflammatory cytokine tumor necrosis factor-alpha (TNF-alpha) is found in ovarian cancer biopsies and is associated with increased tumor grade. In this report, we show that CXCR4 expression on human epithelial ovarian cancer cells is associated with, and can be modulated by, TNF-alpha. Ovarian cancer cells with high endogenous expression of TNF-alpha expressed higher levels of CXCR4 mRNA and protein than cells with low TNF-alpha expression. Stimulation of ovarian cancer cell lines and primary epithelial cancer cells with TNF-alpha resulted in increased CXCR4 mRNA and protein. The TNF-alpha-stimulated increase in CXCR4 mRNA was due partly to de novo synthesis, and up-regulation of CXCR4 cell surface protein increased migration to the CXCR4 ligand CXCL12. CXCR4 mRNA and protein was down-regulated by anti-TNF-alpha antibody or by targeting TNF-alpha mRNA using RNAi. TNF-alpha stimulation activated components of the nuclear factor kappaB pathway, and overexpression of the inhibitor of kappaB also reduced CXCR4 expression. Coculture of macrophages with ovarian cancer cells also resulted in cancer cell up-regulation of CXCR4 mRNA in a TNF-alpha-dependent manner. Finally, there was a correlation between the levels of TNF-alpha and CXCR4 mRNA in clinical biopsies of ovarian cancer, and TNF-alpha protein was expressed in CXCR4-positive tumor cells. TNF-alpha is a critical mediator of tumor promotion in a number of experimental cancers. Our data suggest that one mechanism may be through nuclear factor kappaB-dependent induction of CXCR4.


Subject(s)
Ovarian Neoplasms/metabolism , Receptors, CXCR4/biosynthesis , Tumor Necrosis Factor-alpha/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/physiology , Chemokine CXCL12 , Chemokines, CXC/physiology , Coculture Techniques , Down-Regulation , Female , Humans , I-kappa B Proteins/biosynthesis , Macrophages/cytology , Macrophages/immunology , Macrophages/metabolism , Ovarian Neoplasms/immunology , RNA Interference , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, CXCR4/genetics , Transcription Factors/biosynthesis , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/pharmacology , Up-Regulation
8.
Int J Dev Biol ; 48(5-6): 489-96, 2004.
Article in English | MEDLINE | ID: mdl-15349823

ABSTRACT

Cytokine and chemokine gradients are central to the directed movement of cells in both homeostatic and pathological processes. Most cancers have a complex chemokine network which can influence immune responses to the tumor, direct the extent and cellular composition of the leukocyte infiltrate and also play a role in angiogenesis. Tumor cells can also hijack the chemokine system and gain expression of certain chemokine receptors and respond to specific chemokine gradients. Chemokine receptor expression and activation on malignant cells may be central to the growth, survival and migration of cancer cells from the primary tumor. Chemokine receptors, both CC and CXC have been detected on malignant cells and the relevant ligands are sometimes expressed at the tumor site and at sites of tumor spread, suggesting a role for the chemokine family in malignant growth and metastasis.


Subject(s)
Chemokines/metabolism , Gene Expression Regulation, Neoplastic , Neoplasms/metabolism , Animals , Cell Movement , Chemokine CXCL12 , Chemokines, CXC/metabolism , Disease Progression , Humans , Models, Biological , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasms/pathology , Neovascularization, Pathologic , Receptors, CXCR4/metabolism , Signal Transduction
9.
Cancer Res ; 63(23): 8360-5, 2003 Dec 01.
Article in English | MEDLINE | ID: mdl-14678997

ABSTRACT

The leukocyte infiltrate of human and murine epithelial cancers is regulated by chemokine production in the tumor microenvironment. In this article, we tested the hypothesis that chemokine receptor antagonists may have anticancer activity by inhibiting this infiltrate. We first characterized CC chemokines, chemokine receptors, and the leukocyte infiltrate in the 410.4 murine model of breast cancer. We found that CCL5 (RANTES) was produced by the tumor cells, and its receptors, CCR1 and CCR5, were expressed by the leukocyte infiltrate. As Met-CCL5 is an antagonist of CCR1 and CCR5 with activity in models of inflammatory disease, we tested its activity against 410.4 tumors. After 5 weeks of daily treatment with Met-CCL5, the volume and weight of 410.4 tumors was significantly decreased compared with control-treated tumors. Met-CCL5 was also active against established tumors. The total cell number obtained after collagenase digestion was decreased in Met-CCL5-treated tumors as was the proportion of infiltrating macrophages. Furthermore, chemokine antagonist treatment increased stromal development and necrosis. Our results provide direct evidence that macrophages contribute to tumor development and are the first indication that chemokine receptor antagonists may provide novel strategies in cancer prevention and treatment.


Subject(s)
CCR5 Receptor Antagonists , Mammary Neoplasms, Experimental/pathology , Receptors, Chemokine/antagonists & inhibitors , Animals , Cell Division/drug effects , Chemokine CCL5/biosynthesis , Female , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Macrophages/drug effects , Macrophages/immunology , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/metabolism , Mice , Mice, Inbred BALB C , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, CCR1 , Receptors, CCR5/biosynthesis , Receptors, CCR5/genetics , Receptors, Chemokine/biosynthesis , Receptors, Chemokine/genetics
10.
Cancer Res ; 62(20): 5930-8, 2002 Oct 15.
Article in English | MEDLINE | ID: mdl-12384559

ABSTRACT

Of 14 chemokine receptors investigated, only CXCR4 was expressed on ovarian cancer cells [C. J. Scotton et al., Cancer Res., 61: 4961-4965, 2001]. To further understand the role of this chemokine receptor in ovarian tumor biology, we studied the action of its ligand, CXCL12 (stromal cell-derived factor 1), on the CXCR4-expressing ovarian cancer cell lines IGROV. Ligand stimulation of the CXCR4 receptor resulted in sustained activation of Akt/protein kinase B and biphasic phosphorylation of p44/42 mitogen-activated protein kinase in IGROV. When IGROV cells were cultured under suboptimal conditions, CXCL12 stimulated their in vitro growth, an effect that was abrogated by neutralizing antibodies to CXCR4. This increase in cell number was attributable to stimulation of DNA synthesis, not protection from apoptosis. CXCL12 treatment of IGROV cells also induced mRNA and protein for tumor necrosis factor alpha, a cytokine that is expressed by tumor cells in ovarian cancer biopsies. IGROV cells invaded through Matrigel toward a CXCL12 gradient. Invasion was abrogated by the broad spectrum matrix metalloproteinase and TNFalpha converting enzyme inhibitor Marimastat and was partially inhibited by neutralizing antitumor necrosis factor alpha antibodies. These effects were not limited to the IGROV cell line. They could also be demonstrated in the CAOV-3 ovarian cancer cell line and primary ovarian tumor cells isolated from ovarian ascites. These biological effects of CXCL12 on IGROV cells were also inhibited by the small molecular weight CXCR4 antagonist AMD3100. Finally, we found abundant intracellular CXCL12 protein in tumor cells in 15 of 18 ovarian cancer biopsies but not in epithelial cells from normal ovary or borderline disease. The chemokine CXCL12 may have multiple biological effects in ovarian cancer, stimulating cell migration and invasion through extracellular matrix, as well as DNA synthesis and establishment of a cytokine network in situations that are suboptimal for tumor cell growth.


Subject(s)
Chemokines, CXC/pharmacology , Ovarian Neoplasms/metabolism , Protein Serine-Threonine Kinases , Receptors, CXCR4/physiology , Benzylamines , Cell Division/drug effects , Cell Division/physiology , Chemokine CXCL12 , Cyclams , Enzyme Activation/drug effects , Female , Heterocyclic Compounds/pharmacology , Humans , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Neoplasm Invasiveness , Ovarian Neoplasms/enzymology , Ovarian Neoplasms/pathology , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Receptors, CXCR4/antagonists & inhibitors , Receptors, CXCR4/drug effects , Receptors, CXCR4/metabolism , Tumor Necrosis Factor-alpha/biosynthesis
11.
J Invest Dermatol ; 119(4): 850-7, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12406330

ABSTRACT

The regulation of dendritic cells is far from fully understood. Interestingly, several recent reports have suggested a role for natural killer cells in affecting dendritic cell maturation and function upon direct contact between the cells. It is not known if this interaction takes place also in vivo, or if a potential interaction of natural killer cells and dendritic cells would be affected by allergen exposure of the dendritic cells. The yeast Malassezia can act as an allergen in atopic eczema/dermatitis syndrome, and induce maturation of dendritic cells. Our aims were to study the distribution of natural killer cells in the skin from atopic eczema/dermatitis syndrome patients with the emphasis on possible natural killer cell-dendritic cell interaction, and to assess whether the interaction of Malassezia with dendritic cells would affect subsequent interaction between dendritic cells and natural killer cells. A few scattered natural killer (CD56+/CD3-) cells were found in the dermis of healthy individuals and in nonlesional skin from atopic eczema/dermatitis syndrome patients. In lesional skin and in biopsies from Malassezia atopy-patch-test-positive skin, however, natural killer cells were differentially distributed and for the first time we could show close contact between natural killer cells and CD1a+ dendritic cells. Dendritic cells preincubated with Malassezia became less susceptible to natural-killer-cell-induced cell death, suggesting a direct effect imposed by Malassezia upon interaction of dendritic cells with natural killer cells. These findings indicate that natural killer cells and dendritic cells can interact in the skin and that Malassezia affects the interaction between natural killer cells and dendritic cells. Our data suggest that natural killer cells may play a role in regulating dendritic cells in atopic eczema/dermatitis syndrome.


Subject(s)
Cell Communication , Dendritic Cells/immunology , Dermatitis, Atopic/immunology , Killer Cells, Natural/immunology , Malassezia/physiology , Coculture Techniques , Cytokines/biosynthesis , Cytotoxicity, Immunologic , Humans
12.
Eur J Immunol ; 32(9): 2393-400, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12207323

ABSTRACT

Endothelins (ET-1, ET-2 and ET-3) are 21-amino acid vasoactive peptides that bind to G-protein-linked transmembrane receptors, ET-RA and ET-RB. As well as modulating vasoconstriction, endothelins regulate growth in several cell types and may also affect differentiation, inflammation and angiogenesis. Both macrophages and endothelins are found in areas of hypoxia in solid tumors and ET-2 expression may be modulated by hypoxia in some tumors. As the peptide structure of mature endothelins is similar to that of CXC chemokines, we asked if endothelins contribute to control of macrophage distribution in tumors. We found that ET-2 is a chemoattractant for macrophages and THP-1 monocytic cells, but not for freshly isolated monocytes. The chemotactic response to ET-2 shows a typical bell-shaped response curve. Experiments with endothelin receptor antagonists showed that migration to ET-2 is mediated via the ET-RB receptor. Moreover, monocytes do not express ET-RB. Chemotaxis towards ET-2 is via the MAPK pathway: p44 and p42 are phosphorylated when THP-1 cells are stimulated with ET-2, and the MAPKK inhibitor PD98059 stops chemotaxis. As with 'classical' chemokines, migration toET-2 is also inhibited by hypoxia and by pertussis toxin. As well as its chemotactic properties, ET-2 leads to activation of macrophages. In human breast tumors that express ET-2, endothelins and ET-RB expressing macrophages often co-localized. While shorter than 'classical' chemokines, ET-2 shares a similar peptide sequence with chemokines and may signal via a similar receptor and MAPK-mediated pathway. Furthermore, ET-2 expression by tumors may modulate the behavior of macrophages such that activated cells accumulate in areas of hypoxia.


Subject(s)
Chemotactic Factors/physiology , Chemotaxis/physiology , Endothelin-2/physiology , Macrophages/drug effects , Monocytes/drug effects , Bacterial Proteins/pharmacology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/metabolism , Carcinoma, Ductal, Breast/pathology , Cell Hypoxia , Cell Line , Chemokines, CXC/chemistry , Chemotactic Factors/chemistry , Chemotactic Factors/pharmacology , Chemotaxis/drug effects , Endothelin Receptor Antagonists , Endothelin-1/pharmacology , Endothelin-1/physiology , Endothelin-2/chemistry , Endothelin-2/pharmacology , Enzyme Inhibitors/pharmacology , Female , Flavonoids/pharmacology , Humans , MAP Kinase Signaling System/drug effects , Macrophage Activation/drug effects , Macrophages/physiology , Membrane Proteins/pharmacology , Monocytes/physiology , Neoplasm Proteins/physiology , Oligopeptides , Peptides, Cyclic , Phosphorylation , Piperidines , Protein Processing, Post-Translational/drug effects , Receptor, Endothelin A , Receptor, Endothelin B , Receptors, Endothelin/drug effects , Receptors, Endothelin/physiology , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...