501
|
Piovan E, Tosello V, Indraccolo S, Masiero M, Persano L, Esposito G, Zamarchi R, Ponzoni M, Chieco-Bianchi L, Dalla-Favera R, Amadori A. Differential regulation of hypoxia-induced CXCR4 triggering during B-cell development and lymphomagenesis. Cancer Res 2007; 67:8605-14. [PMID: 17875700 DOI: 10.1158/0008-5472.can-06-4722] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The chemokine receptor CXCR4 plays a central role in organ-specific homing and tumor spreading and is induced by hypoxia. B lymphocytes are exposed to low oxygen tensions during their development, but the influence of hypoxia on their physiology is poorly understood. Here, we show that hypoxia is associated with up-regulation of CXCR4 expression in human normal and malignant B cells, through both transcriptional and posttranslational mechanisms. However, a dichotomic functional response to CXCR4 triggering was observed: both peripheral B cells and lymphomas arising from mature B cells displayed increased responses to CXCR4 triggering under hypoxia, whereas germinal center (GC) B cells as well as GC-derived lymphomas showed CXCR4 receptor desensitization. This phenomenon was associated with differential modulation of key signal-transducing molecules, including mitogen-activated protein kinase phosphatase-1 and regulator of G protein signaling molecule-1. The unresponsiveness of GC-derived lymphomatous B cells to CXCR4 triggering under hypoxia may have implications for the development and pathogenesis of GC-derived lymphoid tumors.
Collapse
MESH Headings
- Animals
- B-Lymphocytes/metabolism
- B-Lymphocytes/pathology
- B-Lymphocytes/physiology
- Burkitt Lymphoma/genetics
- Burkitt Lymphoma/metabolism
- Burkitt Lymphoma/pathology
- Cell Hypoxia/physiology
- Cell Line, Tumor
- Dual Specificity Phosphatase 1/biosynthesis
- Dual Specificity Phosphatase 1/genetics
- Humans
- Hypoxia-Inducible Factor 1, alpha Subunit/genetics
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Lymphoma, B-Cell/genetics
- Lymphoma, B-Cell/metabolism
- Lymphoma, B-Cell/pathology
- Mice
- Mice, SCID
- RGS Proteins/biosynthesis
- RGS Proteins/genetics
- RNA, Small Interfering/genetics
- Receptors, CXCR4/biosynthesis
- Receptors, CXCR4/genetics
- Transcription, Genetic
- Up-Regulation
Collapse
Affiliation(s)
- Erich Piovan
- Oncology Section, Department of Oncology and Surgical Sciences, University of Padova, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
502
|
Trentin L, Miorin M, Facco M, Baesso I, Carraro S, Cabrelle A, Maschio N, Bortoli M, Binotto G, Piazza F, Adami F, Zambello R, Agostini C, Semenzato G. Multiple myeloma plasma cells show different chemokine receptor profiles at sites of disease activity. Br J Haematol 2007; 138:594-602. [PMID: 17686053 DOI: 10.1111/j.1365-2141.2007.06686.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Chemokines and their receptors play a pivotal role in the regulation of B-lymphocyte trafficking. This study was aimed at investigating the pattern of chemokine receptor expression, including CCR1 to CCR3, CCR5 to CCR7, CXCR1 to CXCR5, and the migration ability of multiple myeloma (MM) plasma cells (PC). PC were recovered from the bone marrow (BM) of 29 MM patients, extramedullary sites of 10 patients and the BM of five controls. Flow cytometry analysis showed that the receptors mainly expressed on malignant BM PC were represented by CXCR4 (70% of patients), CCR1 (25%), CCR2 (25%), CCR5 (17%) and CXCR3 (20%), while other receptors were commonly lacking. The analysis performed on extramedullary (peripheral blood and pleural effusion) malignant PC demonstrated that the most represented receptors were CXCR4 (100%), CCR2 (66%) and CXCR1 (60%). The migratory capability of malignant PC at resting conditions identified three groups of patients with different migration (low, intermediate and high). As CXCR4 was the relevant chemokine receptor expressed by MM PC, its ligand CXCL12 induced their migration. These data suggest that malignant PC from MM display different chemokine receptor profiles and that CXCR4 is fully functional and might play a role in the spreading of the disease.
Collapse
Affiliation(s)
- Livio Trentin
- Department of Clinical and Experimental Medicine, Hematology and Clinical Immunology Branch, Padua University School of Medicine, Padua, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
503
|
Koizumi K, Hojo S, Akashi T, Yasumoto K, Saiki I. Chemokine receptors in cancer metastasis and cancer cell-derived chemokines in host immune response. Cancer Sci 2007; 98:1652-8. [PMID: 17894551 PMCID: PMC11159633 DOI: 10.1111/j.1349-7006.2007.00606.x] [Citation(s) in RCA: 169] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The chemotactic cytokines called chemokines are a superfamily of small secreted cytokines that were initially characterized through their ability to prompt the migration of leukocytes. Attention has been focused on the chemokine receptors expressed on cancer cells because cancer cell migration and metastasis show similarities to leukocyte trafficking. CXC chemokine receptor 4 (CXCR4) was first investigated as a chemokine receptor that is associated with lung metastasis of breast cancers. Recently, CXCR4 was reported to be a key molecule in the formation of peritoneal carcinomatosis in gastric cancer. In the present review, we highlight current knowledge about the role of CXCR4 in cancer metastases. In contrast to chemokine receptors expressed on cancer cells, little is known about the roles of cancer cell-derived chemokines. Cancer tissue consists of both cancer cells and various stromal cells, and leukocytes that infiltrate into cancer are of particular importance in cancer progression. Although colorectal cancer invasion is regulated by the chemokine CCL9-induced infiltration of immature myeloid cells into cancer, high-level expression of cancer cell-derived chemokine CXCL16 increases infiltrating CD8(+) and CD4(+) T cells into cancer tissues, and correlates with a good prognosis. We discuss the conflicting biological effects of cancer cell-derived chemokines on cancer progression, using CCL9 and CXCL16 as examples.
Collapse
Affiliation(s)
- Keiichi Koizumi
- Division of Pathogenic Biochemistry, Institute of Natural Medicine, University of Toyama, Toyama 930-0194, Japan.
| | | | | | | | | |
Collapse
|
504
|
Pattarozzi A, Gatti M, Barbieri F, Würth R, Porcile C, Lunardi G, Ratto A, Favoni R, Bajetto A, Ferrari A, Florio T. 17beta-estradiol promotes breast cancer cell proliferation-inducing stromal cell-derived factor-1-mediated epidermal growth factor receptor transactivation: reversal by gefitinib pretreatment. Mol Pharmacol 2007; 73:191-202. [PMID: 17959712 DOI: 10.1124/mol.107.039974] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
The coordinated activity of estrogens and epidermal growth factor receptor (EGFR) family agonists represents the main determinant of breast cancer cell proliferation. Stromal cell-derived factor-1 (SDF-1) enhances extracellular signal-regulated kinases 1 and 2 (ERK1/2) activity via the transactivation of EGFR and 17beta-estradiol (E2) induces SDF-1 production to exert autocrine proliferative effects. On this basis, we evaluated whether the inhibition of the tyrosine kinase (TK) activity of EGFR may control different mitogenic stimuli in breast tumors using the EGFR-TK inhibitor gefitinib to antagonize the proliferation induced by E2 in T47D human breast cancer cells. EGF, E2, and SDF-1 induced a dose-dependent T47D cell proliferation, that being nonadditive suggested the activation of common intracellular pathways. Gefitinib treatment inhibited not only the EGF-dependent proliferation and ERK1/2 activation but also the effects of SDF-1 and E2, suggesting that these activities were mediated by EGFR transactivation. Indeed, both SDF-1 and E2 caused EGFR tyrosine phosphorylation. The molecular link between E2 and SDF-1 proliferative effects was identified because 1,1'-(1,4-phenylenebis(methylene))-bis-1,4,8,11-tetraazacyclotetradecane octahydrochloride (AMD3100), a CXCR4 antagonist, inhibited SDF-1- and E2-dependent proliferation and EGFR and ERK1/2 phosphorylation. EGFR transactivation was dependent on c-Src activation. E2 treatment caused a powerful SDF-1 release from T47D cells. Finally, in SKBR3, E2-resistant cells, EGFR was constitutively activated, and AMD3100 reduced EGFR phosphorylation and cell proliferation, whereas HER2-neu was transactivated by SDF-1 in SKBR3 but not in T47D cells. In conclusion, we show that activation of CXCR4 transduces proliferative signals from the E2 receptor to EGFR, whose inhibition is able to revert breast cancer cell proliferation induced by multiple receptor activation.
Collapse
Affiliation(s)
- Alessandra Pattarozzi
- Sezione Farmacologia, Dipartimento Oncologia, Biologia e Genetica, Università di Genova, Viale Benedetto XV, 2, 16132 Genova, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
505
|
Meier R, Mühlethaler-Mottet A, Flahaut M, Coulon A, Fusco C, Louache F, Auderset K, Bourloud KB, Daudigeos E, Ruegg C, Vassal G, Gross N, Joseph JM. The chemokine receptor CXCR4 strongly promotes neuroblastoma primary tumour and metastatic growth, but not invasion. PLoS One 2007; 2:e1016. [PMID: 17925864 PMCID: PMC1995764 DOI: 10.1371/journal.pone.0001016] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2007] [Accepted: 09/19/2007] [Indexed: 11/19/2022] Open
Abstract
Neuroblastoma (NB) is a heterogeneous, and particularly malignant childhood neoplasm in its higher stages, with a propensity to form metastasis in selected organs, in particular liver and bone marrow, and for which there is still no efficient treatment available beyond surgery. Recent evidence indicates that the CXCR4/CXCL12 chemokine/receptor axis may be involved in promoting NB invasion and metastasis. In this study, we explored the potential role of CXCR4 in the malignant behaviour of NB, using a combination of in vitro functional analyses and in vivo growth and metastasis assessment in an orthotopic NB mouse model. We show here that CXCR4 overexpression in non-metastatic CXCR4-negative NB cells IGR-NB8 and in moderately metastatic, CXCR4 expressing NB cells IGR-N91, strongly increased tumour growth of primary tumours and liver metastases, without altering the frequency or the pattern of metastasis. Moreover shRNA-mediated knock-down experiments confirmed our observations by showing that silencing CXCR4 in NB cells impairs in vitro and almost abrogates in vivo growth. High levels of CXCL12 were detected in the mouse adrenal gland (the primary tumour site), and in the liver suggesting a paracrine effect of host-derived CXCL12 on NB growth. In conclusion, this study reveals a yet unreported NB-specific predominant growth and survival-promoting role of CXCR4, which warrants a critical reconsideration of the role of CXCR4 in the malignant behaviour of NB and other cancers.
Collapse
Affiliation(s)
- Roland Meier
- Department of Paediatrics, Paediatric Oncology Research, University Hospital Centre Hospitalier Universitaire Vaudois Lausanne (CHUV), Lausanne, Switzerland
| | - Annick Mühlethaler-Mottet
- Department of Paediatrics, Paediatric Oncology Research, University Hospital Centre Hospitalier Universitaire Vaudois Lausanne (CHUV), Lausanne, Switzerland
| | - Marjorie Flahaut
- Department of Paediatrics, Paediatric Oncology Research, University Hospital Centre Hospitalier Universitaire Vaudois Lausanne (CHUV), Lausanne, Switzerland
| | - Aurélie Coulon
- Department of Paediatrics, Paediatric Oncology Research, University Hospital Centre Hospitalier Universitaire Vaudois Lausanne (CHUV), Lausanne, Switzerland
| | - Carlo Fusco
- Division of Experimental Pathology, University Institute of Pathology, Lausanne, Switzerland
| | | | - Katya Auderset
- Department of Paediatrics, Paediatric Oncology Research, University Hospital Centre Hospitalier Universitaire Vaudois Lausanne (CHUV), Lausanne, Switzerland
| | - Katia Balmas Bourloud
- Department of Paediatrics, Paediatric Oncology Research, University Hospital Centre Hospitalier Universitaire Vaudois Lausanne (CHUV), Lausanne, Switzerland
| | - Estelle Daudigeos
- Unité Propre de Recherche de l'Enseignement Supérieur EA3535, Institut Gustave Roussy, Villejuif, France
| | - Curzio Ruegg
- Division of Experimental Oncology, Multidisciplinary Oncology Centre (CePO), Lausanne Cancer Centre, Lausanne, Switzerland
| | - Gilles Vassal
- Unité Propre de Recherche de l'Enseignement Supérieur EA3535, Institut Gustave Roussy, Villejuif, France
| | - Nicole Gross
- Department of Paediatrics, Paediatric Oncology Research, University Hospital Centre Hospitalier Universitaire Vaudois Lausanne (CHUV), Lausanne, Switzerland
- * To whom correspondence should be addressed. E-mail:
| | - Jean-Marc Joseph
- Department of Paediatrics, University Hospital Centre Hospitalier Universitaire Vaudois Lausanne (CHUV) Surgery Unit, Lausanne, Switzerland
| |
Collapse
|
506
|
Ben-Baruch A. Organ selectivity in metastasis: regulation by chemokines and their receptors. Clin Exp Metastasis 2007; 25:345-56. [PMID: 17891505 DOI: 10.1007/s10585-007-9097-3] [Citation(s) in RCA: 203] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2007] [Accepted: 09/05/2007] [Indexed: 12/16/2022]
Abstract
Cancer metastasis results from a non-random process, in which organ selectivity by the tumor cells is largely determined by factors that are expressed at the remote organs that eventually turn into preferred sites of metastasis formation. These factors support the consecutive steps required for metastasis formation, including tumor cell adhesion to microvessel walls, extravasation into target tissue and migration. Of the different components that regulate organ selectivity, instrumental roles were recently attributed to chemokines and their receptors. The present review presents the rationale standing behind the first studies looking at the potential involvement of chemokine-related components in organ selectivity. Based on these studies and many others that followed, the current paradigm is that chemokines that are expressed at specific organs determine to large extent organ specificity by promoting tumor cell adhesion to microvessel walls, by facilitating processes of extravasation into the target tissue and by inducing tumor cell migration. Moreover, chemokines can possibly support additional steps that are required for "successful" establishment of metastases, such as tumor cell proliferation and survival. The review focuses on the CXCL12-CXCR4 pair as the role model in our current understanding of chemokine involvement in organ selectivity. This review also describes the prominent roles played by CCR7 and its corresponding chemokine ligands (CCL21, CCL19) in lymph node metastasis, and of the CCR10-CCL27 axis in melanoma skin survival and metastasis. Overall, the present discussion describes chemokines as important constituents of the tumor microenvironment at metastatic sites, dictating directionality of chemokine receptor-expressing tumor cells, facilitating their adhesion and extravasation, and eventually contributing to organ selectivity.
Collapse
Affiliation(s)
- Adit Ben-Baruch
- Cancer Biology Research Center, Department of Cell Research and Immunology, George S Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
507
|
Wendt MK, Cooper AN, Dwinell MB. Epigenetic silencing of CXCL12 increases the metastatic potential of mammary carcinoma cells. Oncogene 2007; 27:1461-71. [PMID: 17724466 DOI: 10.1038/sj.onc.1210751] [Citation(s) in RCA: 93] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Expression of the chemokine receptor CXCR4 has been linked with increased metastasis and decreased clinical prognosis in breast cancer. The current paradigm dictates that CXCR4 fosters carcinoma cell metastasis along a chemotactic gradient to organs expressing the ligand CXCL12. The present study asked if alterations in autocrine CXCR4 signaling via dysregulation of CXCL12 in mammary carcinoma cells modulated their metastatic potential. While CXCR4 was consistently detected, expression of CXCL12 characteristic of human mammary epithelium was silenced by promoter hypermethylation in breast cancer cell lines and primary mammary tumors. Stable re-expression of functional CXCL12 in ligand null cells increased orthotopic primary tumor growth in the mammary fat-pad model of tumorigenesis. Those data parallel increased carcinoma cell proliferation measured in vitro with little-to-no-impact on apoptosis. Moreover, re-expression of autocrine CXCL12 markedly reduced metastatic lung invasion assessed using in vivo bioluminescence imaging following tail vein injection. Consistent with those data, decreased metastasis reflected diminished intracellular calcium signaling and chemotactic migration in response to exogenous CXCL12 independent of changes in CXCR4 expression. Together these data suggest that an elevated migratory signaling response to ectopic CXCL12 contributes to the metastatic potential of CXCR4-expressing mammary carcinoma cells, subsequent to epigenetic silencing of autocrine CXCL12.
Collapse
Affiliation(s)
- M K Wendt
- Department of Microbiology and Molecular Genetics, Medical College of Wisconsin, Milwaukee, WI 53226-0509, USA
| | | | | |
Collapse
|
508
|
Bian XW, Yang SX, Chen JH, Ping YF, Zhou XD, Wang QL, Jiang XF, Gong W, Xiao HL, Du LL, Chen ZQ, Zhao W, Shi JQ, Wang JM. PREFERENTIAL EXPRESSION OF CHEMOKINE RECEPTOR CXCR4 BY HIGHLY MALIGNANT HUMAN GLIOMAS AND ITS ASSOCIATION WITH POOR PATIENT SURVIVAL. Neurosurgery 2007; 61:570-8; discussion 578-9. [PMID: 17881971 DOI: 10.1227/01.neu.0000290905.53685.a2] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVE CXCR4 is implicated in the growth, metastasis, and angiogenesis of malignant tumors. We investigated the potential role of CXCR4 in human gliomas. METHODS The expression of CXCR4 messenger ribonucleic acid and protein by human glioma cell lines was examined by reverse-transcriptase polymerase chain reaction and immunocytochemistry analysis. Tumor cell chemotaxis and production of vascular endothelial growth factor induced by the CXCR4 ligand SDF-1beta were measured. Xenograft models were used for evaluation of glioma cell tumorigenesis. CXCR4 expression by xenografted tumors and primary human glioma specimens were evaluated for CXCR4 protein expression. The relationship between CXCR4 expression and patient survival was analyzed. A synthetic lipoxygenase inhibitor, Nordy, was tested for its effects on glioma cell expression and function of CXCR4, as well as on glioma cell tumorigenicity. RESULTS CXCR4 expression correlated directly with the degree of malignancy of the human glioma cell lines and primary tumors. Activation of CXCR4 induced tumor cell chemotaxis and increased production of vascular endothelial growth factor. Glioma cells expressing higher levels of CXCR4 formed more rapidly growing and lethal tumors in nude mice. Primary human glioma specimens expressing CXCR4 contained high-density microvessels. Patients with CXCR4-positive gliomas had poorer prognosis after surgery. The lipoxygenase inhibitor Nordy diminished CXCR4 expression by glioma cell lines in vitro and reduced their tumorigenicity in nude mice. CONCLUSION The level of CXCR4 expression seems to correlate with the degree of malignancy of human gliomas and may contribute to their rapid growth.
Collapse
Affiliation(s)
- Xiu-wu Bian
- Institute of Pathology, Southwest Hospital, Third Military Medical University, Chongqing, China.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
509
|
Xu C, Sui J, Tao H, Zhu Q, Marasco WA. Human anti-CXCR4 antibodies undergo VH replacement, exhibit functional V-region sulfation, and define CXCR4 antigenic heterogeneity. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2007; 179:2408-18. [PMID: 17675502 DOI: 10.4049/jimmunol.179.4.2408] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The chemokine receptor CXCR4 and its ligand stromal-derived factor-1 (SDF-1/CXCL12) are essential for many biological processes and various pathological conditions. However, the relationship between CXCR4 antigenic structure and SDF-1-mediated biological responses is poorly understood. In this report, a panel of human anti-CXCR4 Abs were isolated and used to explore CXCR4 antigenic heterogeneity and function. Multiple fixed CXCR4 antigenic isoforms were detected on the surface of hemopoietic cells. Epitope mapping studies demonstrated the complex nature of the surface-exposed CXCR4 epitopes. Ab-mediated inhibition of chemotaxis correlated strongly with binding affinity, epitope recognition, as well as the level of CXCR4 isoform expression. In addition, detailed genetic analyses of these Abs showed evidence of V(H) replacement. Importantly, structural and biochemical studies demonstrated tyrosine sulfation in novel regions of the V genes that contributed bidirectionally to the binding activity of the Abs. These data provide the first evidence that functional tyrosine sulfation occurs in self-reactive Abs and suggest a potential new mechanism that may contribute to the pathogenesis of Ab-mediated autoimmune disease. These Abs also provide valuable tools to explore the selective in vivo targeting of CXCR4 isoforms that may be preferentially expressed in certain disease states and involved in steady-state CXCR4-SDF-1 homeostasis.
Collapse
Affiliation(s)
- Chen Xu
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | | | | | | |
Collapse
|
510
|
|
511
|
Abstract
Chemokines are critical mediators of cell migration during routine immune surveillance, inflammation, and development. Chemokines bind to G protein-coupled receptors and cause conformational changes that trigger intracellular signaling pathways involved in cell movement and activation. Although chemokines evolved to benefit the host, inappropriate regulation or utilization of these proteins can contribute to or cause many diseases. Specific chemokine receptors provide the portals for HIV to get into cells, and others contribute to inflammatory diseases and cancer. Thus, there is significant interest in developing receptor antagonists. To this end, the structures of ligands coupled with mutagenesis studies have revealed mechanisms for antagonism based on modified proteins. Although little direct structural information is available on the receptors, binding of small molecules to mutant receptors has allowed the identification of key residues involved in the receptor-binding pockets. In this review, we discuss the current knowledge of chemokine:receptor structure and function, and its contribution to drug discovery.
Collapse
Affiliation(s)
- Samantha J Allen
- Skaggs School of Pharmacy and Pharmaceutical Science, University of California, San Diego, La Jolla, California 92093, USA.
| | | | | |
Collapse
|
512
|
Tsutsumi H, Tanaka T, Ohashi N, Masuno H, Tamamura H, Hiramatsu K, Araki T, Ueda S, Oishi S, Fujii N. Therapeutic potential of the chemokine receptor CXCR4 antagonists as multifunctional agents. Biopolymers 2007; 88:279-89. [PMID: 17167792 DOI: 10.1002/bip.20653] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The chemokine receptor CXCR4 possesses multiple critical functions in normal and pathologic physiology. CXCR4 is a G-protein-coupled receptor that transduces signals of its endogenous ligand, the chemokine CXCL12 (stromal cell-derived factor-1, SDF-1). The interaction between CXCL12 and CXCR4 plays an important role in the migration of progenitors during embryologic development of the cardiovascular, hemopoietic, central nervous systems, and so on. This interaction is also known to be involved in several intractable disease processes, including HIV infection, cancer cell metastasis, leukemia cell progression, rheumatoid arthritis (RA), and pulmonary fibrosis. It is conjectured that this interaction may be a critical therapeutic target in all of these diseases, and several CXCR4 antagonists have been proposed as potential drugs. Fourteen-mer peptides, T140 and its analogues, were previously developed in our laboratory as specific CXCR4 antagonists that were identified as HIV-entry inhibitors, anti-cancer-metastatic agents, anti-chronic lymphocytic/acute lymphoblastic leukemia agents, and anti-RA agents. Cyclic pentapeptides, such as FC131 [cyclo(D-Tyr-Arg-Arg-L-3-(2-naphthyl)alanine-Gly)], were also previously found as CXCR4 antagonist leads based on pharmacophores of T140. This review article describes the elucidation of multiple functions of CXCR4 antagonists and the development of a number of low-molecular weight CXCR4 antagonists involving FC131 analogues and other compounds with different scaffolds including linear-type structures.
Collapse
Affiliation(s)
- Hiroshi Tsutsumi
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
513
|
Arya M, Ahmed H, Silhi N, Williamson M, Patel HRH. Clinical importance and therapeutic implications of the pivotal CXCL12-CXCR4 (chemokine ligand-receptor) interaction in cancer cell migration. Tumour Biol 2007; 28:123-31. [PMID: 17510563 DOI: 10.1159/000102979] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2006] [Accepted: 12/01/2006] [Indexed: 12/20/2022] Open
Abstract
Chemokines are small, secreted proteins and are now the largest known cytokine family. They mediate their effects through a family of G-protein-coupled receptors and were initially recognized for their ability to act as chemo-attractants and activators of specific types of leucocytes in a variety of immune and inflammatory responses. However, during the past 5 years there has been a chemokine revolution in cancer and all scientists and clinicians in oncology-related fields are now aware of their crucial role at all stages of neoplastic transformation and progression. The most important chemokine ligand-receptor interaction is that of the CXCL12 (stromal cell-derived factor-1, SDF-1) ligand with its exclusive receptor CXCR4; this interaction has a pivotal role in the directional migration of cancer cells during the metastatic process. This has been demonstrated by in vitro and in vivo experiments in addition to retrospective clinical studies. These findings have exciting implications in the field of cancer therapeutics, with several small molecule CXCR4 antagonists having been developed, which may provide clinical benefit in the therapy of cancer metastasis. Interestingly, it is likely that the effect of the anti-HER2 antibody [trastuzumab (Herceptin] in breast cancer involves downregulation of the CXCR4 receptor. Unfortunately, a major problem is that chemokine receptors are expressed in other cells within the body, particularly those of the immune system and it is not clear what effects long-term CXCR4 antagonism could have on innate and adaptive immunity. However, there is little doubt that the great strides made in elucidating the complex relationship between chemokines and their role in cancer will soon translate into significant survival benefits for patients.
Collapse
|
514
|
Zhang JP, Lu WG, Ye F, Chen HZ, Zhou CY, Xie X. Study on CXCR4/SDF-1alpha axis in lymph node metastasis of cervical squamous cell carcinoma. Int J Gynecol Cancer 2007; 17:478-83. [PMID: 17362322 DOI: 10.1111/j.1525-1438.2007.00786.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
CXCR4/stromal-cell-derived factor-1alpha (SDF-1alpha) is involved in many cancer metastatic mechanisms. Cervical squamous cell cancer (SCC) tissues (n=35), normal cervical tissues (n=10), metastatic (n=10) and nonmetastatic lymph nodes (n=50), and Hela cells were stained immunohistochemically with CXCR4 monoclonal antibody (mAb). Meanwhile, lymph nodes were stained immunohistochemically with rabbit anti-SDF-1alpha. In vitro invasion of Hela cells was evaluated using Transwell Permeable Supports (Corning, NY), in which Hela cells with/without CXCR4 mAb preincubation were seeded in the upper chambers and medium containing 0-100 ng/mL SDF-1alpha was added to the lower compartments. For evaluating the effect of CXCR4/SDF-1alpha on proliferation of cervical cancer cells, Hela cells were cultured for 72 h exposed to SDF-1alpha with and without CXCR4 mAb. We found that CXCR4 was expressed on SCC cells in all cervical cancer, metastatic lymph node, and Hela cells but not in normal cervix. SDF-1alpha was expressed on lymph cells in all lymph nodes. SDF-1alpha induced the directed migration of Hela cells with a concentration-dependent model, which was inhibited by CXCR4 mAb (P<0.05). SDF-1alpha also stimulated the proliferation of Hela cells mediated by CXCR4 (P<0.05). CXCR4/SDF-1alpha axis probably participates in the metastasis toward lymph nodes in cervical cancer.
Collapse
Affiliation(s)
- J-P Zhang
- Department of Gynecologic Oncology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | | | | | | | | | | |
Collapse
|
515
|
Harvey JR, Mellor P, Eldaly H, Lennard TWJ, Kirby JA, Ali S. Inhibition of CXCR4-mediated breast cancer metastasis: a potential role for heparinoids? Clin Cancer Res 2007; 13:1562-70. [PMID: 17332302 DOI: 10.1158/1078-0432.ccr-06-1987] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE The pattern of breast cancer metastasis may be determined by interactions between CXCR4 on breast cancer cells and CXCL12 within normal tissues. Glycosaminoglycans bind chemokines for presentation to responsive cells. This study was designed to test the hypothesis that soluble heparinoid glycosaminoglycan molecules can disrupt the normal response to CXCL12, thereby reducing the metastasis of CXCR4-expressing cancer cells. EXPERIMENTAL DESIGN Inhibition of the response of CXCR4-expressing Chinese hamster ovary cells to CXCL12 was assessed by measurement of calcium flux and chemotaxis. Radioligand binding was also assessed to quantify the potential of soluble heparinoids to prevent specific receptor ligation. The human breast cancer cell line MDA-MB-231 and a range of sublines were assessed for their sensitivity to heparinoid-mediated inhibition of chemotaxis. A model of hematogenous breast cancer metastasis was established, and the potential of clinically relevant doses of heparinoids to inhibit CXCL12 presentation and metastatic disease was assessed. RESULTS Unfractionated heparin and the low-molecular-weight heparin tinzaparin inhibited receptor ligation and the response of CXCR4-expressing Chinese hamster ovary cells and human breast cancer cell lines to CXCL12. Heparin also removed CXCL12 from its normal site of expression on the surface of parenchymal cells in the murine lung. Both heparin and two clinically relevant dose regimens of tinzaparin reduced hematogenous metastatic spread of human breast cancer cells to the lung in a murine model. CONCLUSIONS Clinically relevant concentrations of tinzaparin inhibit the interaction between CXCL12 and CXCR4 and may be useful to prevent chemokine-driven breast cancer metastasis.
Collapse
Affiliation(s)
- James R Harvey
- Breast Research Group, School of Surgical and Reproductive Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | | | | | | | | | | |
Collapse
|
516
|
Seidl H, Richtig E, Tilz H, Stefan M, Schmidbauer U, Asslaber M, Zatloukal K, Herlyn M, Schaider H. Profiles of chemokine receptors in melanocytic lesions: de novo expression of CXCR6 in melanoma. Hum Pathol 2007; 38:768-80. [PMID: 17306330 DOI: 10.1016/j.humpath.2006.11.013] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2006] [Revised: 11/08/2006] [Accepted: 11/13/2006] [Indexed: 01/09/2023]
Abstract
Selective expression of certain chemokine receptors by melanoma cells and the presence of their ligands in tissues might govern organ site-specific metastasis. Because the expression profile of chemokine receptors in tissues of melanocytic origin is unknown, we performed a comprehensive study on melanocytic tissue samples investigating the expression of 18 chemokine receptors at the mRNA level by real-time polymerase chain reaction, using a semiquantitative approach, and of 3 chemokine receptors (CXCR6, CCR9, and XCR1) at the protein level. We report on the de novo expression of CXCR6 in primary melanomas and melanoma metastases, but absence in melanoma cell lines and congenital nevi. CXCR4 and CCR1 were the only 2 chemokine receptors that were consistently expressed in melanocytes, melanoma cell lines, primary, and metastatic melanoma; CCR1 expression increased significantly over progression. CCR9 and XCR1 transcripts were found in melanocytic lesions, and expression was confirmed by immunohistochemistry. Transcripts for CCR10 were not found in any of the lesions, but in some melanoma cell lines. Expression of CCR7 was observed in primary melanomas and some metastases. CCR5 was exclusively expressed in primary melanomas and some cutaneous metastases. Results revealed a restricted and differential pattern of chemokine receptor expression in melanoma tissue, which varies substantially from the expression profile of melanoma cell lines and warrants functional studies on some receptors.
Collapse
MESH Headings
- Cell Line, Tumor
- Humans
- Immunohistochemistry
- Lymphatic Metastasis
- Melanocytes/metabolism
- Melanoma/metabolism
- Polymerase Chain Reaction
- RNA, Messenger/analysis
- Receptors, CCR
- Receptors, CCR1
- Receptors, CXCR4/metabolism
- Receptors, CXCR5
- Receptors, CXCR6
- Receptors, Chemokine/metabolism
- Receptors, G-Protein-Coupled/metabolism
- Receptors, Virus/metabolism
Collapse
Affiliation(s)
- Hannes Seidl
- Department of Dermatology, Tumor Biology Unit, Medical University of Graz, Graz, Austria
| | | | | | | | | | | | | | | | | |
Collapse
|
517
|
Soon L, Braet F, Condeelis J. Moving in the right direction-nanoimaging in cancer cell motility and metastasis. Microsc Res Tech 2007; 70:252-7. [PMID: 17279509 DOI: 10.1002/jemt.20411] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Although genetic and protein manipulations have been the cornerstone for the study and understanding of biological processes for many decades, complimentary nanoscale observations have only more recently been achieved in the live-imaging mode. It is at the nano measurement level that events such as protein-protein interactions, enzymatic conversions, and single-molecule stochastic behavior take place. Therefore, nanoscale observations allow us to reinterpret knowledge from large-scale or bulk techniques and gain new insight into molecular events that has cellular, tissue, and organismal phenotypic manifestations. This review identifies pertinent questions relating to the sensing and directional component of cancer cell chemotaxis and discusses the platforms that provide insight into the molecular events related to cell motility. The study of cell motility at the molecular imaging level often necessitates the use of devices such as microinjection, microfluidics, in vivo/intravital and in vitro chemotaxis assays, as well as fluorescence methods like uncaging and FRET. The micro- and nanofabricated devices that facilitate these techniques and their incorporation to specialized microscopes such as the multiphoton, AFM, and TIR-FM, for high-resolution imaging comprise the nanoplatforms used to explore the mechanisms of carcinogenesis. In real-time observations, within a milieu of physiological protein concentrations, true states of dynamic and kinetic fluxes can be monitored.
Collapse
Affiliation(s)
- Lilian Soon
- Australian Key Centre for Microscopy and Microanalysis, The University of Sydney, New South Wales, Australia.
| | | | | |
Collapse
|
518
|
Ping YF, Yao XH, Chen JH, Liu H, Chen DL, Zhou XD, Wang JM, Bian XW. The anti-cancer compound Nordy inhibits CXCR4-mediated production of IL-8 and VEGF by malignant human glioma cells. J Neurooncol 2007; 84:21-9. [PMID: 17415525 DOI: 10.1007/s11060-007-9349-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2007] [Accepted: 01/29/2007] [Indexed: 01/08/2023]
Abstract
The chemokine receptor CXCR4 plays an important role in tumor growth, angiogenesis and metastasis. Our previous studies showed that Nordy, a synthetic chiral compound of nordihydroguaiaretic acid, inhibited the growth and angiogenesis of various malignant tumors. In this study we examined the capacity of Nordy to regulate CXCR4-mediated production of angiogenic factors by human glioblastoma cells. We found that Nordy potently inhibited CXCR4 ligand SDF-1-induced production of IL-8 and vascular endothelial cell growth factor, two important angiogenic factors implicated in the progression of malignant tumors. Further study revealed that the effect of Nordy was attributable to its down-regulation of the expression of functional CXCR4 in glioblastoma cells. These results suggest that the anti-cancer activity of Nordy is due, at least in part, to its suppression of the chemokine receptor CXCR4 thus reducing the production of angiogenic factors by tumor cells.
Collapse
Affiliation(s)
- Yi-fang Ping
- Institute of Pathology, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | | | | | | | | | | | | | | |
Collapse
|
519
|
Zhang L, Yeger H, Das B, Irwin MS, Baruchel S. Tissue microenvironment modulates CXCR4 expression and tumor metastasis in neuroblastoma. Neoplasia 2007; 9:36-46. [PMID: 17325742 PMCID: PMC1803034 DOI: 10.1593/neo.06670] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2006] [Revised: 11/27/2006] [Accepted: 11/28/2006] [Indexed: 11/18/2022] Open
Abstract
Neuroblastoma (NB) is derived from intrinsic migratory neural crest cells and has a high potential for distant metastasis. Growing evidence has implicated chemokine receptors, especially CXCR4, which normally control immune and inflammatory cell migration, as having important roles in tumor progression. In this study, we investigated the expression of CXCR4 in eight different NB cell lines and found that CXCR4 expression is dynamically regulated in NB and can be modulated by different tissue stromata. In addition, we demonstrate that IL-5 and IFN-gamma are released from stromal cells and act as differential mediators for CXCR4 expression. We also overexpressed CXCR4 in two NB cell lines, NUB-7 and SK-N-BE(2), and studied the role of CXCR4 in NB metastasis both in vitro and in vivo. In vitro transwell invasion assay showed that CXCR4 overexpression promoted NB cell migration preferentially toward a bone marrow stromal cell-conditioned medium. Using an in vivo xenograft model, CXCR4-overexpressing cells showed an increased incidence of metastasis, most notably bone marrow metastasis. Our studies reveal critical roles for CXCR4 in NB metastasis and provide insights into the regulatory mechanism of chemokine receptors in NB and the importance of the tissue microenvironment in modulating tumor cell behavior.
Collapse
Affiliation(s)
- Libo Zhang
- New Agent and Innovative Therapy Program, The Hospital for Sick Children, Toronto, Canada
- Department of Pediatric Laboratory Medicine, The Hospital for Sick Children, Toronto, Canada
| | - Herman Yeger
- Department of Pediatric Laboratory Medicine, The Hospital for Sick Children, Toronto, Canada
| | - Bikul Das
- New Agent and Innovative Therapy Program, The Hospital for Sick Children, Toronto, Canada
- Department of Pediatric Laboratory Medicine, The Hospital for Sick Children, Toronto, Canada
| | - Meredith S Irwin
- Division of Hematology and Oncology, Department of Pediatrics, The Hospital for Sick Children, Toronto, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, Canada
| | - Sylvain Baruchel
- New Agent and Innovative Therapy Program, The Hospital for Sick Children, Toronto, Canada
- Division of Hematology and Oncology, Department of Pediatrics, The Hospital for Sick Children, Toronto, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, Canada
| |
Collapse
|
520
|
Abstract
Tumour necrosis factor alpha is a member of the TNF/TNFR cytokine superfamily. In common with other family members, TNF-alpha is involved in maintenance and homeostasis of the immune system, inflammation and host defence. However, there is a 'dark side' to this powerful cytokine; it is now clear that, especially in middle and old age, TNF-alpha is involved in pathological processes such as chronic inflammation, autoimmunity and, in apparent contradiction to its name, malignant disease. This article will discuss the involvement of TNF-alpha in the inflammatory network that contributes to all stages of the malignant process, and consider the possibility that TNF-alpha may be a target for cancer therapy.
Collapse
Affiliation(s)
- Frances Balkwill
- Institute of Cancer and the CR-UK Clinical Centre, Barts and The London, Queen Mary's School of Medicine and Dentistry, 3rd Floor, John Vane Science Centre, Charterhouse Square, London, EC1M 6BQ, UK.
| |
Collapse
|
521
|
Abstract
Tumor development and progression are multifactorial processes, regulated by a large variety of intrinsic and microenvironmental factors. A key role in cancer is played by members of the chemokine superfamily. Chemokines and their receptors are expressed by tumor cells and by host cells, in primary tumors and in specific metastatic loci. The effects of chemokines on tumorigenesis are diverse: While some members of the superfamily significantly support this process, others inhibit fundamental events required for tumor establishment and metastasis. The current review describes the multifaceted roles of chemokines in malignancy, addressing four major aspects of their activities: (1) inducing leukocyte infiltration to tumors and regulating immune functions, with emphasis on tumor-associated macrophages (and the chemokines CCL2, CCL5), T cells (and the chemokines CXCL9, CXCL10) and dendritic cells (and the chemokines CCL19, CCL20, CCL21); (2) directing the homing of tumor cells to specific metastatic sites (the CXCL12-CXCR4 axis); (3) regulating angiogenic processes (mainly the ELR(+)-CXC and non-ELR-CXC chemokines); (4) acting directly on the tumor cells to control their malignancy-related functions. Together, these different chemokine functions establish a net of interactions between the tumor cells and their microenvironment, and partly dictate the fate of the malignancy cascade.
Collapse
Affiliation(s)
- A Ben-Baruch
- Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel.
| |
Collapse
|
522
|
Shurin MR, Shurin GV, Lokshin A, Yurkovetsky ZR, Gutkin DW, Chatta G, Zhong H, Han B, Ferris RL. Intratumoral cytokines/chemokines/growth factors and tumor infiltrating dendritic cells: friends or enemies? Cancer Metastasis Rev 2007; 25:333-56. [PMID: 17029028 DOI: 10.1007/s10555-006-9010-6] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The tumor microenvironment consists of a variable combination of tumor cells, stromal fibroblasts, endothelial cells and infiltrating leukocytes, such as macrophages, T lymphocytes, and dendritic cells. A variety of cytokines, chemokines and growth factors are produced in the local tumor environment by different cells accounting for a complex cell interaction and regulation of differentiation, activation, function and survival of multiple cell types. The interaction between cytokines, chemokines, growth factors and their receptors forms a comprehensive network at the tumor site, which is primary responsible for overall tumor progression and spreading or induction of antitumor immune responses and tumor rejection. Although the general thought is that dendritic cells are among the first cells migrating to the tumor site and recognizing tumor cells for the induction of specific antitumor immunity, the clinical relevance of dendritic cells at the site of the tumor remains a matter of debate regarding their role in the generation of successful antitumor immune responses in human cancers. While several lines of evidence suggest that intratumoral dendritic cells play an important role in antitumor immune responses, understanding the mechanisms of dendritic cell/tumor cell interaction and modulation of activity and function of different dendritic cell subtypes at the tumor site is incomplete. This review is limited to discussing the role of intratumoral cytokine network in the understanding immunobiology of tumor-associated dendritic cells, which seems to possess different regulatory functions at the tumor site.
Collapse
Affiliation(s)
- Michael R Shurin
- Department of Pathology, University of Pittsburgh Medical Center and Cancer Institute, Pittsburgh, PA 15213, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
523
|
Redjal N, Chan JA, Segal RA, Kung AL. CXCR4 inhibition synergizes with cytotoxic chemotherapy in gliomas. Clin Cancer Res 2007; 12:6765-71. [PMID: 17121897 DOI: 10.1158/1078-0432.ccr-06-1372] [Citation(s) in RCA: 104] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE The chemokine receptor CXCR4 is expressed in many different cancers. In malignant brain tumors, CXCR4 signaling has been implicated in tumor growth, survival, and migration, and pharmacologic inhibition of CXCR4 results in decreased tumor growth in preclinical models. To understand how CXCR4 inhibitors may be incorporated into clinical therapy, we examined determinants of responsiveness to CXCR4 inhibition. Because optimal use of CXCR4 inhibition will likely be a part of multimodality therapy, we also investigated the efficacy of CXCR4 inhibition combined with conventional cytotoxic chemotherapy. EXPERIMENTAL DESIGN CXCR4 protein levels and responsiveness to the CXCR4 inhibitor AMD3100 were determined in a panel of glioblastoma multiforme cell lines. The effects of AMD3100, alone or in combination with 1,3-bis(2-chloroethyl)-1-nitrosourea (BCNU), on cell growth were determined for several of these cell lines in vitro. We used an orthotopic model of glioblastoma multiforme to evaluate the antitumor efficacy of AMD3100 combined with BCNU in vivo. RESULTS The level of CXCR4 protein expression in glioblastoma multiforme cells predicts the dose at which there is a response to AMD3100; cells that express higher levels of CXCR4 protein require higher doses for equivalent response. In all cell lines tested, treatment of glioblastoma multiforme cells with BCNU followed by AMD3100 results in synergistic antitumor efficacy in vitro. This synergy can also be seen in an orthotopic glioblastoma multiforme model. Treatment with subtherapeutic doses of BCNU in combination with AMD3100 results in tumor regression in vivo, and this reflects both increased apoptosis and decreased proliferation following combination therapy. CONCLUSION These studies support testing CXCR4 inhibitors in patients with glioblastoma multiforme and establish that inhibition of CXCR4 synergizes with conventional cytotoxic therapies in a clinically relevant combinatorial strategy.
Collapse
Affiliation(s)
- Navid Redjal
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | | | | | | |
Collapse
|
524
|
Kaplan RN, Psaila B, Lyden D. Niche-to-niche migration of bone-marrow-derived cells. Trends Mol Med 2007; 13:72-81. [PMID: 17197241 DOI: 10.1016/j.molmed.2006.12.003] [Citation(s) in RCA: 175] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2006] [Revised: 11/24/2006] [Accepted: 12/18/2006] [Indexed: 02/06/2023]
Abstract
During ontogenesis, haematopoietic stem cells (HSCs) relocate between extra-embryonic and embryonic compartments. Similarly, site-specific homing of HSCs is ongoing during adulthood. With the expanding knowledge of HSC physiology, a new paradigm emerges in which HSCs and haematopoietic progenitor cells (HPCs) migrate to defined microenvironments within the bone marrow (BM) and to 'activated' or 'inducible' niches elsewhere. Here, we summarize current understanding of HSC niche characteristics, and the physiological and pathological mechanisms that guide HSC homing both within the BM and to distant niches in the periphery, promoting new vessel growth in tumours and ischaemia. Recent observations suggest that features of the HSC niche might also be recapitulated in pre-metastatic sites. Clusters of BM-derived HPCs promote invasion of disseminating cancer cells. Clear clinical benefits can be foreseen by modulating HSCs and their microenvironments, in promoting tissue regeneration, and inhibiting tumourigenesis and cancer metastasis.
Collapse
Affiliation(s)
- Rosandra N Kaplan
- Department of Pediatrics, Weill College of Medicine at Cornell University and Memorial Sloan-Kettering Cancer Center, New York, NY10021, USA
| | | | | |
Collapse
|
525
|
Kodama J, Kusumoto T, Seki N, Matsuo T, Ojima Y, Nakamura K, Hongo A, Hiramatsu Y. Association of CXCR4 and CCR7 chemokine receptor expression and lymph node metastasis in human cervical cancer. Ann Oncol 2007; 18:70-76. [PMID: 17032700 DOI: 10.1093/annonc/mdl342] [Citation(s) in RCA: 98] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The chemokine receptors CXCR4 and CCR7 have been suggested to play an important role in cancer invasion and metastasis. The expression of these receptors in human cervical cancer, however, has seldom been characterized. PATIENTS AND METHODS We investigated the expression of CXCR4 and CCR7 in cervical cancer specimens and determined the association between their expression and the clinicopathological features observed, including patient outcome. RESULTS CXCR4 expression was significantly higher in elderly patients (P=0.025); it was also significantly increased in patients with cancers displaying large tumor size (P=0.010), deep stromal invasion (P=0.0004), lymph-vascular space involvement (P=0.0002), or lymph node metastasis (P<0.0001). CCR7 expression was significantly higher in cases of squamous cell carcinomas (P=0.010) and in patients with cancers showing large tumor size (P<0.0001), deep stromal invasion (P<0.0001), vaginal invasion (P=0.047), lymph-vascular space involvement (P=0.012), or lymph node metastasis (P<0.0001). Logistic regression analysis revealed that deep stromal invasion (P=0.017) and CXCR4 (P=0.016) and CCR7 (P=0.022) expression were independent factors that influenced pelvic lymph node metastasis. The disease-free survival and overall survival (OS) rates of patients exhibiting both CXCR4 and CCR7 expression were significantly reduced (P<0.0001). In addition, the expression of both CXCR4 and CCR7 was an independent prognostic factor for OS (95% confidence interval=1.03-17.86; P=0.046). CONCLUSIONS CXCR4 and CCR7 expression may be associated with lymph node metastasis; moreover, the expression of these receptors can serve as an indicator of poor prognosis in patients with cervical cancer.
Collapse
Affiliation(s)
- J Kodama
- Department of Obstetrics and Gynecology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan.
| | - T Kusumoto
- Department of Obstetrics and Gynecology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - N Seki
- Department of Obstetrics and Gynecology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - T Matsuo
- Department of Obstetrics and Gynecology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Y Ojima
- Department of Obstetrics and Gynecology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - K Nakamura
- Department of Obstetrics and Gynecology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - A Hongo
- Department of Obstetrics and Gynecology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Y Hiramatsu
- Department of Obstetrics and Gynecology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| |
Collapse
|
526
|
Wang Z, Ma Q. β-Catenin is a promising key factor in the SDF-1/CXCR4 axis on metastasis of pancreatic cancer. Med Hypotheses 2007; 69:816-20. [PMID: 17379424 DOI: 10.1016/j.mehy.2007.01.069] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2007] [Accepted: 01/18/2007] [Indexed: 11/30/2022]
Abstract
Metastasis is recently the most fearsome of cancer. Pancreatic cancer is the fourth leading cause of cancer death. Morbidity and mortality from pancreatic cancer is conspicuously associated with metastasis. However, the mechanism of metastasis is not well described. Early studies mostly focus on the "soil and seed" hypothesis. Recently, the chemotaxis hypothesis has been paid more attention. Cancer cell with high expression of chemokine receptor will spread to the specific sites where the ligand is highly secreted. It has been demonstrated that SDF-1/CXCR4 signaling, one of the most important chemokine receptor-ligand complexes, was considered to play a critical role in pancreatic cancer organ-specific metastasis through some possible pathways. However, studies do not clarify the mechanism of SDF-1/CXCR4 signaling on pancreatic cancer progression. Beta-catenin, an important factor in canonical Wnt signaling pathway, also makes great contributions on cancer invasion and metastasis. It seems that Wnt/beta-catenin has a significant role in pancreatic cancer progression through interactions with different protein complexes. In the previous study of neural development, the relationship between SDF-1/CXCR4 signaling and beta-catenin has been described. It gave a clue to describe the correlation between SDF-1/CXCR4 signaling and Wnt/beta-catenin pathway. According to this, we postulate that beta-catenin is a promising key factor of SDF-1/CXCR4 signaling to regulate the metastasis of pancreatic cancer. With the stimulation of SDF-1 on highly metastatic pancreatic cancer cells, beta-catenin will separate from different complexes, translocate into the nucleus, trigger the expression of target genes and finally promote the migration of pancreatic cancer cells to specific sites. Through the observation of this crosstalk, it is possible to understand more clearly about the pancreatic cancer specific metastasis and to make some contributions on gene therapy of pancreatic cancer.
Collapse
Affiliation(s)
- Zheng Wang
- Department of Hepatobillary Surgery, The First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Shaanxi, 710061, PR China
| | | |
Collapse
|
527
|
|
528
|
Massa A, Casagrande S, Bajetto A, Porcile C, Barbieri F, Thellung S, Arena S, Pattarozzi A, Gatti M, Corsaro A, Robello M, Schettini G, Florio T. SDF-1 Controls Pituitary Cell Proliferation through the Activation of ERK1/2 and the Ca2+-Dependent, Cytosolic Tyrosine Kinase Pyk2. Ann N Y Acad Sci 2006; 1090:385-98. [PMID: 17384283 DOI: 10.1196/annals.1378.042] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Stromal cell-derived factor-1 (SDF-1) is a chemokine of the CXC subfamily that exerts its effects via CXCR4, a G-protein-coupled receptor. CXCR4 is often expressed by tumor cells, and its activation causes tumor cell proliferation. Using GH4C1 cells, here we show that SDF-1 induced cell proliferation in a dose-dependent manner. Thus, we evaluated the intracellular signaling involved in this effect. SDF-1 increased cytosolic [Ca2+] and activated Pyk2, ERK1/2, and BKCa channels. To correlate these intracellular effectors with the proliferative activity of SDF-1, we inhibited their activity using BAPTA-AM (Ca2+ chelator), PD98059 (MEK inhibitor), salicylate (Pyk2 inhibitor), and TEA (K+ channel blocker). All these compounds reverted SDF-1-induced proliferation, suggesting the involvement of multiple intracellular pathways. To identify a possible crosstalk and a molecular ordering among these pathways, we tested these antagonists on SDF-1-dependent activation of ERK1/2, Pyk2, and BKCa channels. We report that the inhibition of [Ca2+]i increase or the blockade of BKCa channel activity did not affect ERK1/2 activation by SDF-1; Pyk2 activation was purely Ca2+-dependent, not involving ERK1/2 or BKCa channels; and BKCa channel activity was antagonized by Pyk2 but not by ERK1/2 inhibitors. These data suggest that SDF-1-dependent increase of [Ca2+]i activates Pyk2, which, in turn, regulates BKCa channel activity. Conversely, ERK1/2 activation is an independent phenomenon. In conclusion, we demonstrate that SDF-1 induces proliferation of GH4C1 cells, suggesting that the activation of CXCR4 may represent a novel regulatory mechanism for pituitary cell proliferation which may contribute to pituitary adenoma development.
Collapse
Affiliation(s)
- Alessandro Massa
- Section of Pharmacology, Department of Oncology, Biology, and Genetics, University of Genova, Viale Benedetto XV, 2, 16132 Genova, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
529
|
Bajetto A, Barbieri F, Pattarozzi A, Dorcaratto A, Porcile C, Ravetti JL, Zona G, Spaziante R, Schettini G, Florio T. CXCR4 and SDF1 expression in human meningiomas: a proliferative role in tumoral meningothelial cells in vitro. Neuro Oncol 2006; 9:3-11. [PMID: 17108064 PMCID: PMC1828107 DOI: 10.1215/15228517-2006-023] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Chemokines participate in cellular processes associated with tumor proliferation, migration, and angiogenesis. We previously demonstrated that stromal cell-derived factor 1 (SDF1) exerts a mitogenic activity in glioblastomas through the activation of its receptor CXCR4. Here we studied the expression of this chemokine in human meningiomas and its possible role in cell proliferation. Reverse transcriptase-PCR analysis for CXCR4 and SDF1 was performed on 55 human meningiomas (47 WHO grade I, 5 WHO II, and 3 WHO III). Immunolabeling for CXCR4 and SDF1 was performed on paraffin-embedded sections of these tumors. [(3)H]Thymidine uptake and Western blot analyses were performed on primary meningeal cell cultures of tumors to evaluate the proliferative activity of human SDF1alpha (hSDF1alpha) in vitro and the involvement of extracellular signal-regulated kinase 1/2 (ERK1/2) activation in this process. CXCR4 mRNA was expressed by 78% of the tumor specimens and SDF1 mRNA by 53%. CXCR4 and SDF1 were often detected in the same tumor tissues and colocalized with epithelial membrane antigen immunostaining. In 9 of 12 primary cultures from meningiomas, hSDF1alpha induced significant cell proliferation that was strongly reduced by the mitogen-activated protein kinase kinase inhibitor PD98059, involving ERK1/2 activation in the proliferative signal of hSDF1alpha. In fact, CXCR4 stimulation led to ERK1/2 phosphorylation/activation. In addition, the hSDF1alpha-induced cell proliferation was significantly correlated with the MIB1 staining index in the corresponding surgical specimen. In conclusion, we found that human meningiomas express CXCR4 and SDF1 and that hSDF1alpha induces proliferation in primary meningioma cell cultures through the activation of ERK1/2.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Blotting, Western
- Carcinoma/genetics
- Carcinoma/metabolism
- Carcinoma/pathology
- Cell Proliferation
- Chemokine CXCL12
- Chemokines, CXC/genetics
- Chemokines, CXC/metabolism
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- Immunoenzyme Techniques
- In Vitro Techniques
- Male
- Meningeal Neoplasms/genetics
- Meningeal Neoplasms/metabolism
- Meningioma/genetics
- Meningioma/metabolism
- Meningioma/pathology
- Middle Aged
- Mitogen-Activated Protein Kinase 1/metabolism
- Mitogen-Activated Protein Kinase 3/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- RNA, Neoplasm/genetics
- RNA, Neoplasm/metabolism
- Receptors, CXCR4/genetics
- Receptors, CXCR4/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Signal Transduction
- Stromal Cells/metabolism
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Adriana Bajetto
- Department of Oncology, Biology and Genetics, University of Genova, Viale Benedetto XV, 2, 16132 Genova, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
530
|
Busillo JM, Benovic JL. Regulation of CXCR4 signaling. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2006; 1768:952-63. [PMID: 17169327 PMCID: PMC1952230 DOI: 10.1016/j.bbamem.2006.11.002] [Citation(s) in RCA: 467] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2006] [Accepted: 11/04/2006] [Indexed: 12/31/2022]
Abstract
The chemokine receptor CXCR4 belongs to the large superfamily of G protein-coupled receptors, and is directly involved in a number of biological processes including organogenesis, hematopoiesis, and immune response. Recent evidence has highlighted the role of CXCR4 in a variety of diseases including HIV, cancer, and WHIM syndrome. Importantly, the involvement of CXCR4 in cancer metastasis and WHIM syndrome appears to be due to dysregulation of the receptor leading to enhanced signaling. Herein we review what is currently known regarding the regulation of CXCR4 and how dysregulation contributes to disease progression.
Collapse
Affiliation(s)
- John M. Busillo
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, PA 19107
| | - Jeffrey L. Benovic
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, PA 19107
- To whom correspondence should be addressed:
| |
Collapse
|
531
|
Pan J, Mestas J, Burdick MD, Phillips RJ, Thomas GV, Reckamp K, Belperio JA, Strieter RM. Stromal derived factor-1 (SDF-1/CXCL12) and CXCR4 in renal cell carcinoma metastasis. Mol Cancer 2006; 5:56. [PMID: 17083723 PMCID: PMC1636662 DOI: 10.1186/1476-4598-5-56] [Citation(s) in RCA: 141] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2006] [Accepted: 11/03/2006] [Indexed: 12/31/2022] Open
Abstract
Renal cell carcinoma (RCC) is characterized by organ-specific metastases. The chemokine stromal derived factor-1 (SDF-1/CXCL12) and its receptor CXCR4 have been suggested to regulate organ-specific metastasis in various other cancers. On this basis, we hypothesized that the biological axis of CXCL12 via interaction with its receptor, CXCR4, is a major mechanism for RCC metastasis. We demonstrated that CXCR4 was significantly expressed on circulating cytokeratin+ RCC cells from patients with known metastatic RCC. We detected up-regulation of CXCR4 mRNA and protein levels on a human RCC cell line by either knockdown of the von Hippel-Lindau (VHL) tumor suppressor protein, or incubating the cells under hypoxic conditions. The enhanced CXCR4 expression was mediated through the interaction of the Hypoxia Inducible Factor-1α (HIF-1α) with the promoter region of the CXCR4 gene. Furthermore, the expression of CXCR4 on human RCC directly correlated with their metastatic ability in vivo in both heterotopic and orthotopic SCID mouse models of human RCC. Neutralization of CXCL12 in SCID mice abrogated metastasis of RCC to target organs expressing high levels of CXCL12; without altering tumor cell proliferation, apoptosis, or tumor-associated angiogenesis. Therefore, our data suggest that the CXCL12/CXCR4 biological axis plays an important role in regulating the organ-specific metastasis of RCC.
Collapse
MESH Headings
- Animals
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Carcinoma, Renal Cell/genetics
- Carcinoma, Renal Cell/metabolism
- Carcinoma, Renal Cell/secondary
- Cell Hypoxia
- Cell Line, Tumor
- Chemokine CXCL12
- Chemokines, CXC/antagonists & inhibitors
- Chemokines, CXC/pharmacology
- Chemokines, CXC/physiology
- Chemotaxis/drug effects
- Gene Expression Regulation, Neoplastic
- Humans
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Keratins/analysis
- Kidney Neoplasms/genetics
- Kidney Neoplasms/metabolism
- Kidney Neoplasms/pathology
- Mice
- Mice, SCID
- Promoter Regions, Genetic
- RNA Interference
- Receptors, CXCR4/genetics
- Receptors, CXCR4/metabolism
- Transcriptional Activation
- Von Hippel-Lindau Tumor Suppressor Protein/antagonists & inhibitors
- Von Hippel-Lindau Tumor Suppressor Protein/genetics
Collapse
Affiliation(s)
- Judong Pan
- Department of Medicine, David Geffen School of Medicine at UCLA, 900 Veteran Ave., Los Angeles, California, USA
| | - Javier Mestas
- Department of Medicine, David Geffen School of Medicine at UCLA, 900 Veteran Ave., Los Angeles, California, USA
| | - Marie D Burdick
- Department of Medicine, David Geffen School of Medicine at UCLA, 900 Veteran Ave., Los Angeles, California, USA
| | - Roderick J Phillips
- Department of Medicine, David Geffen School of Medicine at UCLA, 900 Veteran Ave., Los Angeles, California, USA
| | - George V Thomas
- Department of Pathology, David Geffen School of Medicine at UCLA, 900 Veteran Ave., Los Angeles, California, USA
| | - Karen Reckamp
- Department of Medicine, David Geffen School of Medicine at UCLA, 900 Veteran Ave., Los Angeles, California, USA
| | - John A Belperio
- Department of Medicine, David Geffen School of Medicine at UCLA, 900 Veteran Ave., Los Angeles, California, USA
| | - Robert M Strieter
- Department of Medicine, David Geffen School of Medicine at UCLA, 900 Veteran Ave., Los Angeles, California, USA
- Department of Pathology, David Geffen School of Medicine at UCLA, 900 Veteran Ave., Los Angeles, California, USA
- Department of Pediatrics, David Geffen School of Medicine at UCLA, 900 Veteran Ave., Los Angeles, California, USA
| |
Collapse
|
532
|
Wald O, Izhar U, Amir G, Avniel S, Bar-Shavit Y, Wald H, Weiss ID, Galun E, Peled A. CD4+CXCR4highCD69+ T Cells Accumulate in Lung Adenocarcinoma. THE JOURNAL OF IMMUNOLOGY 2006; 177:6983-90. [PMID: 17082613 DOI: 10.4049/jimmunol.177.10.6983] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The chemokine receptor CXCR4 is involved in the growth and metastasis of tumor cells. However, the expression of its ligand, the chemokine CXCL12, in tumors and its role in regulating the accumulation of immune cells within the tumors is not clear. Using ELISA and immunohistochemistry we found that CXCL12 is expressed in the majority of nonsmall cell lung cancer tissue sections obtained from stage IA to IIB nonsmall cell lung cancer patients undergoing operation. Histopathologic examination of these sections indicated that high CXCL12 expression correlated with increased tumor inflammation. In addition, disease recurrence rates in a subgroup of adenocarcinoma patients showed a tendency to correlate with high CXCL12 expression in the tumor. Isolation of adenocarcinoma-infiltrating immune cells demonstrated an increase in the percentage of CD4+CD69+CXCR4+ T cells as compared with normal lung tissue. About 30% of these cells expressed the regulatory T cell markers CD25high and FoxP3. The percentage of CD8 T cells within the tumor did not change, however; the percentage of NK and NK T cells was significantly reduced. In correlation with CXCR4 expression, CD4 T cells showed increased migration in response to CXCL12 compared with CD8 T cells and NK cells. Overall, these observations suggest that CXCL12 expression may influence tumor progression by shaping the immune cell population infiltrating lung adenocarcinoma tumors.
Collapse
MESH Headings
- Adenocarcinoma/chemistry
- Adenocarcinoma/immunology
- Adenocarcinoma/pathology
- Aged
- Antigens, CD/biosynthesis
- Antigens, Differentiation, T-Lymphocyte/biosynthesis
- Biomarkers, Tumor/biosynthesis
- Biomarkers, Tumor/chemistry
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- CD4-Positive T-Lymphocytes/pathology
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- CD8-Positive T-Lymphocytes/pathology
- Carcinoma, Non-Small-Cell Lung/chemistry
- Carcinoma, Non-Small-Cell Lung/immunology
- Carcinoma, Non-Small-Cell Lung/pathology
- Cell Movement/immunology
- Chemokine CXCL12
- Chemokines, CXC/metabolism
- Female
- Humans
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- Killer Cells, Natural/pathology
- Lectins, C-Type
- Lung Neoplasms/chemistry
- Lung Neoplasms/immunology
- Lung Neoplasms/pathology
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Lymphocytes, Tumor-Infiltrating/pathology
- Male
- Receptors, CXCR4/biosynthesis
- Receptors, CXCR4/metabolism
Collapse
Affiliation(s)
- Ori Wald
- Goldyne Savad Institute of Gene Therapy, Hadassah University Hospital, Jerusalem, Isreal
| | | | | | | | | | | | | | | | | |
Collapse
|
533
|
Scala S, Giuliano P, Ascierto PA, Ieranò C, Franco R, Napolitano M, Ottaiano A, Lombardi ML, Luongo M, Simeone E, Castiglia D, Mauro F, De Michele I, Calemma R, Botti G, Caracò C, Nicoletti G, Satriano RA, Castello G. Human melanoma metastases express functional CXCR4. Clin Cancer Res 2006; 12:2427-33. [PMID: 16638848 DOI: 10.1158/1078-0432.ccr-05-1940] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE The chemokine receptor CXCR4 was identified as an independent predictor of poor prognosis in primary melanoma. The aim of the study was to investigate the role of CXCR4 in human melanoma metastases. EXPERIMENTAL DESIGN CXCR4 expression was evaluated in melanoma metastases and in metastatic cell lines through immunohistochemistry, immunoblotting, immunofluorescence, and reverse transcription-PCR. The function of CXCR4 was tested in the presence of the ligand, CXCL12, through induction of extracellular signal-regulated kinase-1 and -2 (Erk-1 and -2) phosphorylation, proliferation, apoptosis, and migration capabilities. RESULTS CXCR4 expression was detected in 33 out of 63 (52.4%) metastases from cutaneous melanomas. Metastatic melanoma cell lines expressed cell surface CXCR4; PES 43, Alo 40, and COPA cell lines showed the highest levels of CXCR4 (>90% of positive cells); PES 41, Alo 39, PES 47, POAG, and CIMA cell lines showed low to moderate degrees of expression (5-65% of positive cells). Other chemokine receptors, CCR7 and CCR10, were detected on the melanoma cell lines; CXCL12 activated Erk-1 and Erk-2, the whose induction was specifically inhibited by AMD3100 treatment. CXCL12 increased the growth in PES 41, PES 43, and PES 47 cells under suboptimal (1% serum) and serum-free culture conditions; AMD3100 (1 mumol/L) inhibited the spontaneous and CXCL12-induced proliferation. No rescue from apoptosis was shown but PES 41, PES 43, and PES 47 cells migrate toward CXCL12. CONCLUSIONS These findings indicate that CXCR4 is expressed and active in human melanoma metastases, suggesting that active inhibitors such as AMD3100 may be experienced in human melanoma.
Collapse
Affiliation(s)
- Stefania Scala
- Department of Clinical Immunology, National Cancer Institute, G. Pascale Foundation, Naples, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
534
|
Richard CL, Tan EY, Blay J. Adenosine upregulates CXCR4 and enhances the proliferative and migratory responses of human carcinoma cells to CXCL12/SDF-1alpha. Int J Cancer 2006; 119:2044-53. [PMID: 16823836 DOI: 10.1002/ijc.22084] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The level of expression of the chemokine receptor CXCR4 has been shown to play a crucial role in determining the ability of cancer cells to metastasize from the primary tumor and become established in tissue sites that are rich in the CXCR4 ligand CXCL12/SDF-1alpha. High CXCR4 expression on cancer cells is associated with an increased risk of recurrence and poorer overall survival. We propose that local tissue mediators within the primary tumor or at secondary sites may modulate the level of CXCR4 expression and, therefore, potentially affect the ability of the cancer cells to metastasize. The purine nucleoside adenine-9-beta-D-ribofuranoside (adenosine) is generated at high concentrations within the extracellular fluid of solid tumors because of their hypoxia. We show here that adenosine acts through A(2A) and A(2B) adenosine receptors on human colorectal carcinoma cells to upregulate CXCR4 mRNA expression up to 10-fold and selectively increases cell-surface CXCR4 protein up to 3-fold. This increase in cell-surface CXCR4 enables the carcinoma cells to migrate toward CXCL12, and enhances their proliferation in response to CXCL12. Adenosine may therefore be one of the factors within the tumor microenvironment that facilitates tumor dissemination, by upregulating CXCR4 on certain cancer cells and enhancing cellular responses to CXCL12.
Collapse
Affiliation(s)
- Cynthia Lee Richard
- Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| | | | | |
Collapse
|
535
|
Slettenaar VIF, Wilson JL. The chemokine network: a target in cancer biology? Adv Drug Deliv Rev 2006; 58:962-74. [PMID: 16996642 DOI: 10.1016/j.addr.2006.03.012] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2006] [Accepted: 07/10/2006] [Indexed: 01/30/2023]
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.
Collapse
Affiliation(s)
- Violet I F Slettenaar
- Centre for Translational Oncology, Institute of Cancer and the CR-UK Clinical Centre, Barts and The London Queen Mary's School of Medicine and Dentistry, 3rd Floor, John Vane Science Centre, Charterhouse Square, London, EC1M 6BQ, UK
| | | |
Collapse
|
536
|
Slagsvold T, Marchese A, Brech A, Stenmark H. CISK attenuates degradation of the chemokine receptor CXCR4 via the ubiquitin ligase AIP4. EMBO J 2006; 25:3738-49. [PMID: 16888620 PMCID: PMC1553197 DOI: 10.1038/sj.emboj.7601267] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2005] [Accepted: 07/12/2006] [Indexed: 12/13/2022] Open
Abstract
HER2 overexpression in cancers causes hyperactivation of the PI 3-kinase pathway and elevated levels of the chemokine receptor CXCR4, which is strongly associated with increased metastatic potential. Here, we provide evidence that the cytokine-independent survival kinase CISK is activated downstream of the PI 3-kinase-dependent kinase PDK1 on endosomes and negatively regulates the lysosomal degradation of CXCR4. We demonstrate that CISK prevents CXCR4 degradation by inhibiting sorting of the receptor from early endosomes to lysosomes. In contrast, CISK does not interfere with ligand-induced degradation of epidermal growth factor receptors. CISK strongly interacts and colocalizes with the E3 ubiquitin ligase AIP4, which is important for the ubiquitin-dependent lysosomal degradation of CXCR4. Moreover, the observed inhibition is both dependent on the interaction between CISK and AIP4 and on the activation status of CISK. Consistent with this, an activated form of CISK but not of the related kinase SGK1 phosphorylates specific sites of AIP4 in vitro. Taken together, these results reveal a critical function of CISK in specifically attenuating ubiquitin-dependent degradation of CXCR4, and provide a mechanistic link between the PI 3-kinase pathway and CXCR4 stability.
Collapse
Affiliation(s)
- Thomas Slagsvold
- Department of Biochemistry, The Norwegian Radium Hospital and the University of Oslo, Montebello, Oslo, Norway
| | - Adriano Marchese
- Department of Pharmacology, Loyola University Chicago, Stritch School of Medicine, Maywood, IL, USA
- Department of Pharmacology, Loyola University Chicago, Stritch School of Medicine, Maywood, IL 60153, USA. Tel.: +1 708 216 3456; Fax: +1 708 216 6596; E-mail:
| | - Andreas Brech
- Department of Biochemistry, The Norwegian Radium Hospital and the University of Oslo, Montebello, Oslo, Norway
| | - Harald Stenmark
- Department of Biochemistry, The Norwegian Radium Hospital and the University of Oslo, Montebello, Oslo, Norway
- Department of Biochemistry, The Norwegian Radium Hospital and The University of Oslo, Montebello, Oslo 0310, Norway. Tel.: +47 2293 4951; Fax: +47 2250 8692; E-mail:
| |
Collapse
|
537
|
Ueda Y, Neel NF, Schutyser E, Raman D, Richmond A. Deletion of the COOH-terminal domain of CXC chemokine receptor 4 leads to the down-regulation of cell-to-cell contact, enhanced motility and proliferation in breast carcinoma cells. Cancer Res 2006; 66:5665-75. [PMID: 16740704 PMCID: PMC2664111 DOI: 10.1158/0008-5472.can-05-3579] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The CXC chemokine receptor 4 (CXCR4) contributes to the metastasis of human breast cancer cells. The CXCR4 COOH-terminal domain (CTD) seems to play a major role in regulating receptor desensitization and down-regulation. We expressed either wild-type CXCR4 (CXCR4-WT) or CTD-truncated CXCR4 (CXCR4-DeltaCTD) in MCF-7 human mammary carcinoma cells to determine whether the CTD is involved in CXCR4-modulated proliferation of mammary carcinoma cells. CXCR4-WT-transduced MCF-7 cells (MCF-7/CXCR4-WT cells) do not differ from vector-transduced MCF-7 control cells in morphology or growth rate. However, CXCR4-DeltaCTD-transduced MCF-7 cells (MCF-7/CXCR4-DeltaCTD cells) exhibit a higher growth rate and altered morphology, potentially indicating an epithelial-to-mesenchymal transition. Furthermore, extracellular signal-regulated kinase (ERK) activation and cell motility are increased in these cells. Ligand induces receptor association with beta-arrestin for both CXCR4-WT and CXCR4-DeltaCTD in these MCF-7 cells. Overexpressed CXCR4-WT localizes predominantly to the cell surface in unstimulated cells, whereas a significant portion of overexpressed CXCR4-DeltaCTD resides intracellularly in recycling endosomes. Analysis with human oligomicroarray, Western blot, and immunohistochemistry showed that E-cadherin and Zonula occludens are down-regulated in MCF-7/CXCR4-DeltaCTD cells. The array analysis also indicates that mesenchymal marker proteins and certain growth factor receptors are up-regulated in MCF-7/CXCR4-DeltaCTD cells. These observations suggest that (a) the overexpression of CXCR4-DeltaCTD leads to a gain-of-function of CXCR4-mediated signaling and (b) the CTD of CXCR4-WT may perform a feedback repressor function in this signaling pathway. These data will contribute to our understanding of how CXCR4-DeltaCTD may promote progression of breast tumors to metastatic lesions.
Collapse
Affiliation(s)
- Yukiko Ueda
- Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Nicole F. Neel
- Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Evemie Schutyser
- Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Dayanidhi Raman
- Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Ann Richmond
- Departments of Veterans Affairs, Vanderbilt University School of Medicine, Nashville, Tennessee
- Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, Tennessee
| |
Collapse
|
538
|
Fricker SP, Anastassov V, Cox J, Darkes MC, Grujic O, Idzan SR, Labrecque J, Lau G, Mosi RM, Nelson KL, Qin L, Santucci Z, Wong RSY. Characterization of the molecular pharmacology of AMD3100: a specific antagonist of the G-protein coupled chemokine receptor, CXCR4. Biochem Pharmacol 2006; 72:588-96. [PMID: 16815309 DOI: 10.1016/j.bcp.2006.05.010] [Citation(s) in RCA: 162] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2006] [Revised: 05/13/2006] [Accepted: 05/17/2006] [Indexed: 11/28/2022]
Abstract
The chemokine receptor CXCR4 is widely expressed on different cell types, is involved in leukocyte chemotaxis, and is a co-receptor for HIV. AMD3100 has been shown to be a CXCR4 receptor antagonist, and to block HIV infection of T-tropic, X4-using, virus in vitro and in vivo. AMD3100 is an effective mobilizer of hematopoietic stem cells and is being investigated in clinical trials in multiple myeloma and non-Hodgkins lymphoma patients. Using the CCRF-CEM T-cell line that constitutively expresses CXCR4 we confirmed that AMD3100 was an antagonist of SDF-1/CXCL12 ligand binding (IC50=651+/-37 nM). We have also shown that AMD3100 inhibits SDF-1 mediated GTP-binding (IC50=27+/-2.2 nM), SDF-1 mediated calcium flux (IC50=572+/-190 nM), and SDF-1 stimulated chemotaxis (IC50=51+/-17 nM). AMD3100 did not inhibit calcium flux against cells expressing CXCR3, CCR1, CCR2b, CCR4, CCR5 or CCR7 when stimulated with their cognate ligands, nor did it inhibit receptor binding of LTB4. AMD3100 did not, on its own, induce a calcium flux in the CCRF-CEM cells, which express multiple GPCRs including CXCR4, CCR4 and CCR7. Furthermore, AMD3100 neither stimulated GTP-binding, an assay for GPCR activation, in CEM cell membranes; nor chemotaxis of CCRF-CEM cells. These data therefore demonstrate that AMD3100 is a specific antagonist of CXCR4, is not cross-reactive with other chemokine receptors, and is not an agonist of CXCR4.
Collapse
Affiliation(s)
- Simon P Fricker
- AnorMED Inc., #200 20353 64th Avenue, Langley, BC V2Y 1N5, Canada.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
539
|
Tamamura H, Fujii N. The therapeutic potential of CXCR4 antagonists in the treatment of HIV infection, cancer metastasis and rheumatoid arthritis. Expert Opin Ther Targets 2006; 9:1267-82. [PMID: 16300475 DOI: 10.1517/14728222.9.6.1267] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
CXCR4 is the receptor of the chemokine CXCL12, which is involved in progression and metastasis of several types of cancer cells, HIV infection and rheumatoid arthritis. The authors developed selective CXCR4 antagonists, T22 and T140, initially as anti-HIV agents, which inhibit T cell line-tropic (X4-) HIV-1 infection through their specific binding to CXCR4. Recently, T140 analogues have also been shown to inhibit CXCL12-induced migration of breast cancer cells, leukaemia T cells, pancreatic cancer cells, small cell lung cancer cells, chronic lymphocytic leukaemia B cells, pre-B acute lymphoblastic leukaemia cells and so on in vitro. Biostable T140 analogues significantly suppressed pulmonary metastasis of breast cancer cells and melanoma cells in mice. Furthermore, these compounds significantly suppressed the delayed-type hypersensitivity response induced by sheep red blood cells and collagen-induced arthritis, which represent in vivo mouse models of arthritis. Thus, T140 analogues proved to be attractive lead compounds for chemotherapy of these problematic diseases. This article reviews recent research on T140 analogues, referring to several other CXCR4 antagonists.
Collapse
Affiliation(s)
- Hirokazu Tamamura
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo 101-0062, Japan.
| | | |
Collapse
|
540
|
Dewan MZ, Ahmed S, Iwasaki Y, Ohba K, Toi M, Yamamoto N. Stromal cell-derived factor-1 and CXCR4 receptor interaction in tumor growth and metastasis of breast cancer. Biomed Pharmacother 2006; 60:273-6. [PMID: 16828253 DOI: 10.1016/j.biopha.2006.06.004] [Citation(s) in RCA: 153] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2006] [Indexed: 11/23/2022] Open
Abstract
Stromal cell-derived factor-1 (SDF-1)/CXCR4 interaction is critical for the trafficking of lymphocytes, homing and retention of hematopoietic stem cells within the bone marrow and is essential in fetal hematopoiesis. Binding of SDF-1 to CXCR4 activates a variety of intracellular signal transduction pathways and effector molecules that regulate cell survival, proliferation, chemotaxis, migration and adhesion. Recently, intensive research has demonstrated that SDF-1/CXCR4 interaction also regulates several key events in wide variety of cancers. Serum-depleted media in the presence of SDF-1 protected the breast cancer cells from apoptosis. CXCR4-low-expressing MCF-7 formed small tumor at inoculated site in SCID mice 8-9 weeks after inoculation while completely failed to metastasis into various organs. In contrast, CXCR4-high-expressing MDA-231 cells were most efficient in the formation of a large tumor and organ-metastasis within 3 weeks in SCID mice. This review briefly focuses on the role of SDF-1/CXCR4 interaction in tumor growth and metastasis of breast cancer cell both in vitro and in vivo.
Collapse
Affiliation(s)
- M Z Dewan
- Department of Molecular Virology, Bio-Response, Graduate School, Tokyo Medical and Dental University, 1-5-45 Yushima, Tokyo 113-8519, Japan
| | | | | | | | | | | |
Collapse
|
541
|
Miyazaki H, Patel V, Wang H, Edmunds RK, Gutkind JS, Yeudall WA. Down-regulation of CXCL5 inhibits squamous carcinogenesis. Cancer Res 2006; 66:4279-84. [PMID: 16618752 DOI: 10.1158/0008-5472.can-05-4398] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We report a novel role for the CXC-chemokine, CXCL5, in the proliferation and invasion of head and neck squamous cell carcinoma (HNSCC). Previously, we reported transcriptional up-regulation of CXCL5 in metastatic cells. In this study, we provide biological validation of these findings and show that CXCL5 is intimately involved in tumor cell proliferation, migration, and invasion. Cells derived from a lymph node metastasis, but not from a synchronous primary tumor, secreted CXCL5 as judged by Western blotting of conditioned media. We used RNA interference to generate cell lines (shL5) in which CXCL5 expression was greatly reduced, and tested whether this modulated the cell phenotype. shL5 cells showed decreased proliferation compared with cells harboring nontargeting control sequences. In addition, we found that the ability of shL5 cells to migrate and invade in vitro through a basement membrane substitute was greatly impaired compared with control cells. Finally, whereas control cells were highly tumorigenic in nude mice, the tumorigenic potential in vivo of shL5 cells was found to be ablated. Taken together, these data suggest that CXCL5 production contributes to both enhanced proliferation and invasion of squamous cell carcinomas and that targeting of chemokine pathways may represent a potential therapeutic modality for these lesions.
Collapse
MESH Headings
- Carcinoma, Squamous Cell/genetics
- Carcinoma, Squamous Cell/metabolism
- Carcinoma, Squamous Cell/pathology
- Cell Growth Processes/physiology
- Cell Line, Tumor
- Cell Movement/physiology
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/metabolism
- Chemokine CXCL5
- Chemokines, CXC/antagonists & inhibitors
- Chemokines, CXC/biosynthesis
- Chemokines, CXC/genetics
- Disease Progression
- Down-Regulation
- Head and Neck Neoplasms/genetics
- Head and Neck Neoplasms/metabolism
- Head and Neck Neoplasms/pathology
- Humans
- Neoplasm Invasiveness
- Neoplasm Metastasis
- RNA Interference
- RNA, Small Interfering/genetics
- Transfection
Collapse
Affiliation(s)
- Hiroshi Miyazaki
- Philips Institute, Virginia Commonwealth University, Richmond, Virginia 23298-0566, USA
| | | | | | | | | | | |
Collapse
|
542
|
Oppenheimer SB. Cellular basis of cancer metastasis: A review of fundamentals and new advances. Acta Histochem 2006; 108:327-34. [PMID: 16730054 DOI: 10.1016/j.acthis.2006.03.008] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2006] [Revised: 03/15/2006] [Accepted: 03/21/2006] [Indexed: 12/12/2022]
Abstract
This review provides an introduction to fundamentals and new advances in cancer metastasis for general readers. The first segment includes topics such as cell adhesion, cell migration, proteases, inflammation, coagulation and site selection in metastasis. Then follows a discussion of an interesting report by Kaplan et al. [VEGFR1-positive haematopoietic bone marrow progenitors initiate the pre-metastatic niche. Nature 2005;438:820-7] that provides evidence for a role of VEGFR1+bone marrow cells in preparing pre-metastatic niches in specific organs that host the arrival and growth of metastatic cancer cells. The therapeutic implications of this study are explored.
Collapse
Affiliation(s)
- Steven B Oppenheimer
- Department of Biology and Center for Cancer Developmental Biology, California State University Northridge, 18111 Nordhoff Street, Northridge, CA 91330-8303, USA.
| |
Collapse
|
543
|
Zhu YM, Bagstaff SM, Woll PJ. Production and upregulation of granulocyte chemotactic protein-2/CXCL6 by IL-1beta and hypoxia in small cell lung cancer. Br J Cancer 2006; 94:1936-41. [PMID: 16721367 PMCID: PMC2361351 DOI: 10.1038/sj.bjc.6603177] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Small cell lung cancer (SCLC) is characterised by early and widespread metastasis. However, SCLC cells have so far been found to produce low levels of known pro-angiogenic factors. We speculated that SCLC cells might produce alternative pro-angiogenic factors. Here, we report that a panel of SCLC cell lines constitutively secrete granulocyte chemotactic protein-2 (GCP-2)/CXCL6, a CXC ELR+ chemokine. In contrast, none of the three tested NSCLC cell lines secreted GCP-2. Production of GCP-2 in vivo was also confirmed in seven out of nine specimens with SCLC. We demonstrate that expression of GCP-2 is mediated by NF-kappaB as ALLN, an NF-kappaB pathway inhibitor, almost completely abolished GCP-2 production in SCLC cell lines. We also demonstrate that GCP-2 can be significantly upregulated by IL-1beta and hypoxia in SCLC cell lines. This result suggests a role for GCP-2 in promoting tumour progression in vivo under unfavourable conditions such as oxygen deprivation. As SCLC cells express both GCP-2 and its receptors CXCR1 and CXCR2, their biological significance in SCLC progression was further studied. We demonstrate that GCP-2 is an autocrine growth factor. Cell proliferation was significantly inhibited by anti-GCP-2 neutralising antibody in two high-GCP-2-producing cell lines. In addition, expression of the proliferation marker PCNA was upregulated by exogenous GCP-2 in two low-GCP-2-producing cell lines. Taken together, these results suggest an important role for GCP-2 as an autocrine mitogen in the growth and metastasis of SCLC.
Collapse
Affiliation(s)
- Y M Zhu
- Department of Clinical Oncology, Division of Genomic Medicine, School of Medicine and Biomedical Sciences, Institute for Cancer Studies, University of Sheffield, UK.
| | | | | |
Collapse
|
544
|
Holt JE, Jackson A, Roman SD, Aitken RJ, Koopman P, McLaughlin EA. CXCR4/SDF1 interaction inhibits the primordial to primary follicle transition in the neonatal mouse ovary. Dev Biol 2006; 293:449-60. [PMID: 16545793 DOI: 10.1016/j.ydbio.2006.02.012] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2005] [Revised: 02/01/2006] [Accepted: 02/13/2006] [Indexed: 11/18/2022]
Abstract
The molecular mechanisms behind the entry of the primordial follicle into the growing follicle pool remain poorly understood. To investigate this process further, a microarray-based comparison was undertaken between 2-day postpartum mouse ovaries consisting of primordial follicles/naked oocytes only and those with both primordial follicles and newly activated follicles (7-day postpartum). Gene candidates identified included the chemoattractive cytokine stromal derived factor-1 (SDF1) and its receptor CXCR4. SDF1 and CXCR4 have been implicated in a variety of physiological processes including the migration of embryonic germ cells to the gonads. SDF1-alpha expression increased with the developmental stage of the follicle. Embryonic expression was found to be dichotomous post-germ cell migration, with low expression in the female. Immunohistochemical studies nonetheless indicate that the autocrine pattern of expression ligand and receptor begins during embryonic life. Addition of recombinant SDF1-alpha to neonatal mouse ovaries in vitro resulted in significantly higher follicle densities than for control ovaries. TUNEL analysis indicated no detectable difference in populations of apoptotic cells of treated or control ovaries. Treated ovaries also contained a significantly lower percentage of activated follicles as determined by measurement of oocyte diameter and morphological analysis. Treatment of cultured ovaries with an inhibitor of SDF1-alpha, AMD3100, ablated the effect of SDF1-alpha. By retaining follicles in an unactivated state, SDF1/CXCR4 signaling may play an important role in maintaining the size and longevity of the primordial follicle pool.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Base Sequence
- Benzylamines
- Chemokine CXCL12
- Chemokines, CXC/antagonists & inhibitors
- Chemokines, CXC/genetics
- Chemokines, CXC/pharmacology
- Chemokines, CXC/physiology
- Cyclams
- DNA, Complementary/genetics
- Female
- Heterocyclic Compounds/pharmacology
- Male
- Mice
- Oocytes/drug effects
- Oocytes/growth & development
- Oocytes/metabolism
- Ovarian Follicle/drug effects
- Ovarian Follicle/embryology
- Ovarian Follicle/growth & development
- Ovarian Follicle/metabolism
- Ovary/drug effects
- Ovary/embryology
- Ovary/growth & development
- Ovary/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptors, CXCR4/antagonists & inhibitors
- Receptors, CXCR4/genetics
- Receptors, CXCR4/physiology
- Recombinant Proteins/pharmacology
- Testis/embryology
- Testis/growth & development
- Testis/metabolism
Collapse
Affiliation(s)
- Janet E Holt
- Reproductive Science Group, Discipline of Biological Science, School of Environmental and Life Sciences, University of Newcastle, Callaghan, NSW 2308, Australia
| | | | | | | | | | | |
Collapse
|
545
|
Expression of CXC chemokine receptors 1-5 and their ligands in human glioma tissues: role of CXCR4 and SDF1 in glioma cell proliferation and migration. Neurochem Int 2006; 49:423-32. [PMID: 16621164 DOI: 10.1016/j.neuint.2006.03.003] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2006] [Accepted: 03/02/2006] [Indexed: 01/01/2023]
Abstract
Chemokines have been involved in cellular processes associated to malignant transformation such as proliferation, migration and angiogenesis. The expression of five CXC chemokine receptors and their main ligands was analysed by RT-PCR in 31 human astrocytic neoplasms. The mRNAs for all the receptors analysed were identified in a high percentage of tumours, while their ligands showed lower expression. CXCR4 and SDF1 were the most frequently mRNA identified (29/31 and 13/31 of the gliomas studied, respectively). Thus, we further analysed the cell localization of CXCR4 and SDF1 in immunohistochemistry experiments. We show a marked co-localization of CXCR4 and SDF1 in tumour cells, mainly evident in psudolpalisade and microcystic degeneration areas and in the vascular endothelium. In addition, hSDF1alpha induced a significant increase of DNA synthesis in primary human glioblastoma cell cultures and chemotaxis in a glioblastoma cell line. These results provide evidence of the expression of multiple CXC chemokines and their receptors in brain tumours and that in particular CXCR4 and SDF1 sustain proliferation and migration of glioma cells to promote malignant progression.
Collapse
|
546
|
Holland JD, Kochetkova M, Akekawatchai C, Dottore M, Lopez A, McColl SR. Differential Functional Activation of Chemokine Receptor CXCR4 Is Mediated by G Proteins in Breast Cancer Cells. Cancer Res 2006; 66:4117-24. [PMID: 16618732 DOI: 10.1158/0008-5472.can-05-1631] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
CXCR4 is a G protein-coupled receptor of considerable biological significance, and among its numerous functions, it is suggested to play a critical role in cancer metastasis. We have investigated the expression and function of CXCR4 in a range of breast cancer cell lines covering a spectrum of invasive phenotypes and found that, while surface levels of CXCR4 were uniform across the entire panel, only highly invasive cells that are metastatic in immunocompromised mice expressed functional receptors. CXCL12/SDF-1 induced cellular responses such as calcium mobilization, actin polymerization, and chemotaxis in metastatic cells, whereas noninvasive cells were unresponsive. Moreover, CXCL12 activated multiple signaling pathways downstream of G proteins in highly invasive cells but failed to activate any of the examined kinase cascades in noninvasive cell lines. This blockade in nonmetastatic cell lines seems to be due to the inability of G protein alpha and beta subunits to form a heterotrimeric complex with CXCR4. Galpha and Gbeta were able to bind to CXCR4 independently in all cell lines, but the association of G protein alphabetagamma heterotrimers with the receptor, a prerequisite for signal transduction downstream from G protein-coupled receptors, was only observed in the highly invasive cell lines. Our findings show, for the first time, that CXCR4 function is subject to complex and potentially tightly controlled regulation in breast cancer cells via differential G protein-receptor complex formation, and this regulation may play a role in the transition from nonmetastatic to malignant tumors.
Collapse
Affiliation(s)
- Jane D Holland
- School of Molecular and Biomedical Science, University of Adelaide, Adelaide, Australia
| | | | | | | | | | | |
Collapse
|
547
|
Wang D, Wang H, Brown J, Daikoku T, Ning W, Shi Q, Richmond A, Strieter R, Dey SK, DuBois RN. CXCL1 induced by prostaglandin E2 promotes angiogenesis in colorectal cancer. ACTA ACUST UNITED AC 2006; 203:941-51. [PMID: 16567391 PMCID: PMC2118273 DOI: 10.1084/jem.20052124] [Citation(s) in RCA: 271] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Chronic inflammation is a well-known risk factor for cancer. Proinflammatory mediators such as prostaglandin E2 (PGE2) promote colorectal tumor growth by stimulating angiogenesis, cell invasion, and cell growth, and inhibiting apoptosis. Molecules that regulate tumor-associated angiogenesis provide promising therapeutic targets for treatment of colorectal cancer (CRC) as indicated by the recent development of the novel anti-angiogenic agent bevacizumab (Avastin). However, use of this drug only prolongs survival by several months, highlighting the importance of finding more effective treatment regimens. We report here that PGE2 induces expression of CXCL1 (growth-regulated oncogene alpha), a pro-angiogenic chemokine, in human CRC cells. More importantly, CXCL1 released from carcinoma cells induces microvascular endothelial cell migration and tube formation in vitro. Furthermore, PGE2 promotes tumor growth in vivo by induction of CXCL1 expression, which results in increased tumor microvessel formation. These results have potential clinical significance because we found that CXCL1 expression correlates with PGE2 levels in human CRCs. Collectively, our findings show for the first time that CXCL1 is regulated by PGE2 and indicate that CXCL1 inhibitors should be evaluated further as potential anti-angiogenic agents for treatment of CRC.
Collapse
MESH Headings
- Adenoma/blood supply
- Adenoma/pathology
- Animals
- Caco-2 Cells
- Cell Line, Tumor
- Cell Movement/immunology
- Chemokine CXCL1
- Chemokines, CXC/biosynthesis
- Chemokines, CXC/genetics
- Chemokines, CXC/metabolism
- Chemokines, CXC/physiology
- Colorectal Neoplasms/blood supply
- Colorectal Neoplasms/pathology
- Dinoprostone/physiology
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/pathology
- ErbB Receptors/physiology
- Female
- Humans
- Intercellular Signaling Peptides and Proteins/biosynthesis
- Intercellular Signaling Peptides and Proteins/genetics
- Intercellular Signaling Peptides and Proteins/metabolism
- Intercellular Signaling Peptides and Proteins/physiology
- Male
- Mice
- Mice, Knockout
- Mice, SCID
- Mice, Transgenic
- Mitogen-Activated Protein Kinases/physiology
- Neovascularization, Pathologic/immunology
- Neovascularization, Pathologic/metabolism
- Receptors, Interleukin-8B/biosynthesis
- Receptors, Interleukin-8B/genetics
Collapse
Affiliation(s)
- Dingzhi Wang
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
548
|
Growth and activation of PI-3K/PKB and Akt by stromal cell-derived factor 1α in endometrial carcinoma cells with expression of suppressor endoprotein PTEN. Chin Med J (Engl) 2006. [DOI: 10.1097/00029330-200603010-00005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
549
|
Woerner BM, Warrington NM, Kung AL, Perry A, Rubin JB. Widespread CXCR4 activation in astrocytomas revealed by phospho-CXCR4-specific antibodies. Cancer Res 2006; 65:11392-9. [PMID: 16357147 DOI: 10.1158/0008-5472.can-05-0847] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The chemokine receptor CXCR4 is expressed in many cancers where it may regulate tumor cell growth and migration. The role of CXCR4 in cancer will depend on it being in an activated, signaling state. To better define the significance of CXCR4 expression in cancer, we developed an antibody that can distinguish CXCR4 phosphorylated on serine 339, a residue previously identified as a site for ligand-induced phosphorylation. With this antibody, we investigated the mechanisms of CXCR4 phosphorylation and evaluated the phosphorylation status of CXCR4 in human astrocytomas. In vitro, phosphorylation of serine 339 occurred in response to CXCL12 or epidermal growth factor (EGF) treatment and was increased by protein kinase C activation. In all grades of astrocytomas, CXCR4 was expressed in tumor cells and some endothelial cells, whereas CXCL12 was present in endothelial cells and infiltrating microglia. We found that CXCR4 phosphorylated on serine 339 was present in tumor cells and vascular endothelial cells in all grades of astrocytoma. These data indicate that CXCR4 is expressed and activated in astrocytomas and that phosphorylation of CXCR4 can occur through ligand activation or transactivation via the EGF receptor. These studies extend the potential roles of CXCR4 in cancer to include functions associated with benign (grade 1) tumors. The ability to distinguish phosphorylated CXCR4 will be invaluable for the continued analysis of the role of CXCR4 in cancer and the development of CXCR4 antagonist therapy for patients suffering with primary tumors of the brain and other sites.
Collapse
Affiliation(s)
- B Mark Woerner
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | | | | | |
Collapse
|
550
|
Coopman PJ, Mueller SC. The Syk tyrosine kinase: a new negative regulator in tumor growth and progression. Cancer Lett 2006; 241:159-73. [PMID: 16442709 DOI: 10.1016/j.canlet.2005.11.004] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2005] [Revised: 11/03/2005] [Accepted: 11/04/2005] [Indexed: 11/28/2022]
Abstract
The spleen tyrosine kinase Syk was long thought to be a hematopoietic cell-specific signaling molecule. Recent evidence demonstrated that it is also expressed by many non-hematopoietic cell types and that it plays a negative role in cancer. A significant drop in its expression was first observed during breast cancer progression, but an anomalous Syk expression has now also been evidenced in many other tumor types. Mechanistic studies using Syk re-expression demonstrated its suppressive function in tumorigenesis and metastasis formation, which is surprising for a tyrosine kinase. Loss of Syk expression is regulated, albeit not exclusively, by its promoter hypermethylation. The molecular mechanism of its tumor-suppressive function remains largely unknown; the identification of its activators and effectors in non-hematopoietic cells will be a challenge for the years to come. An increasing number of clinical studies reveal a correlation between reduced Syk expression and an increased risk for metastasis formation, and assign Syk as a potential new prognostic marker in different tumor types.
Collapse
Affiliation(s)
- Peter J Coopman
- CNRS UMR 5539, Université Montpellier 2, 34095 Montpellier, France.
| | | |
Collapse
|