51
|
Liu H, Yang Y, Xiao J, Yang S, Liu Y, Kang W, Li X, Zhang F. Semaphorin 4D expression is associated with a poor clinical outcome in cervical cancer patients. Microvasc Res 2014; 93:1-8. [PMID: 24603190 DOI: 10.1016/j.mvr.2014.02.007] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Revised: 02/20/2014] [Accepted: 02/20/2014] [Indexed: 01/13/2023]
Abstract
Lymphangiogenesis is thought to be essential for cancer progression, making it an important target in cancer therapy. Lymphangiogenic factors (VEGF-C and VEGF-D) are upregulated in various tumors/cancers, and play an important role in lymphangiogenesis and lymph node metastasis. Similarly, semaphorin 4D (Sema4D) is a potent inducer of angiogenesis, and its overexpression is associated with tumor progression and poor prognosis in a variety of malignancies. However, little is known regarding the functional relationship between Sema4D and VEGF-C/VEGF-D and in the mediation of lymphangiogenesis and lymph node metastasis and clinical outcome. The current study aimed to evaluate the effect of Sema4D expression on outcome in patients with cervical cancer, and to explore the molecular mechanism of Sema4D in tumor progression. We evaluated Sema4D expression, density of lymphatic vessels, and invasion of lymphatic vessels with immunohistochemical methods in 232 human cervical cancers with long-term follow-up. Sema4D expression was correlated with patho-clinical parameters and patients' outcome. A cervical cancer cell line was used to investigate the contribution of sema4D to tumor progression by studying the role of Sema4D in VEGF-C/-D and cell migration using reverse transcription-polymerase chain reaction and Western blotting. We observed that Sema4D expression was higher in metastatic cervical cancer than in nonmetastatic cervical cancer (P<0.001). CD34-positive or D2-40-positive lymphatic vessel density was significantly increased in cases with lymph node metastasis compared with those without lymph node metastasis. The increased Sema4D expression was associated with VEGF-C/-D, the presence of lymphatic invasion, the occurrence of lymph node metastasis, and FIGO stage. We also observed a novel association between Sema4D upregulation and poor prognosis in cervical cancer. In vitro, the Sema4D inhibitory antibody and Sema4D-shRNA suppressed VEGF-C and VEGF-D in the human cervical carcinoma cell lines HeLa, Siha, and Caski cells. Invasiveness assay demonstrated that Sema4D could augment the invasive potential of the tumor cells in the cervical cancer lines and induction of cellular invasiveness by Sema4D stimulation could be inhibited by knockdown of plexinB1 by siRNA. Further mechanistic investigations of tumor cell invasiveness showed that Sema4D could induce activation of GTPase Ras homolog gene family, member A (RhoA), MAPK and AKT. In addition, plexinB1 knockdown by siRNA could suppress the Sema4D signal transmitted to MAPK and Akt. Taken together, these results suggest that Sema4D autocrine within tumor cells contributes to enhanced invasion and tumor progression through increased motility of cervical cancer and VEGF-C/-D-mediated lymphangiogenesis. Sema4D might be useful as a molecular marker of poor prognosis in cervical cancer.
Collapse
Affiliation(s)
- Huidong Liu
- Department of Anatomy, Harbin Medical University, 194 Xuefu Road, 150081 Harbin, China
| | - Yanmei Yang
- Cancer Institute, Harbin Medical University, 150 Harping Road, 150081 Harbin, China
| | - Jianbing Xiao
- Department of Anatomy, Harbin Medical University, 194 Xuefu Road, 150081 Harbin, China
| | - Shucai Yang
- Department of Anatomy, Harbin Medical University, 194 Xuefu Road, 150081 Harbin, China
| | - Yan Liu
- Department of General Surgery, Mudanjiang First Hospital, 4 Jianwei Road, 157011 Mudanjiang, China
| | - Wenhui Kang
- Department of Radiotherapy, The Third Affiliated Hospital of Harbin Medical University, 150 Harping Road, 150081 Harbin, China
| | - Xinlei Li
- Department of Anatomy, Harbin Medical University, 194 Xuefu Road, 150081 Harbin, China
| | - Fengmin Zhang
- Department of Medical Microbiology, Heilongjiang Province Key Laboratory for Immunity and Infection, Harbin Medical University, 194 Xuefu Road, 150081 Harbin, China.
| |
Collapse
|
52
|
Cazes A, Gibault L, Rivera C, Mordant P, Riquet M. [Lymphatic extension and lymphangiogenesis in non-small cell lung cancer]. REVUE DE PNEUMOLOGIE CLINIQUE 2014; 70:26-31. [PMID: 24566036 DOI: 10.1016/j.pneumo.2013.09.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Accepted: 09/23/2013] [Indexed: 06/03/2023]
Abstract
Lymph node metastasis is a major adverse prognostic factor of malignant tumors, including non-small cell lung carcinoma (NSCLC). However the characterization of tumor associated lymphatic vessels and lymphangiogenic mediators in NSCLC are recent and their prognostic role is debated. Lymphatic vascular invasion (LVI) appears like a robust adverse prognostic factor when reported in NSCLC. This parameter should be better standardized and could be of use in adjuvant therapy indications. Moreover, anti-lymphangiogenesis therapies are currently under investigation and may become part of the anti-cancer strategy.
Collapse
Affiliation(s)
- A Cazes
- Service d'anatomie pathologique, hôpital européen Georges-Pompidou, 20, rue Leblanc, 75015 Paris, France; Unversité Paris Descartes, 12, rue de l'École-de-Médecine, 75006 Paris, France
| | - L Gibault
- Service d'anatomie pathologique, hôpital européen Georges-Pompidou, 20, rue Leblanc, 75015 Paris, France
| | - C Rivera
- Service de chirurgie thoracique, hôpital européen Georges-Pompidou, 20, rue Leblanc, 75015 Paris, France; Unversité Paris Descartes, 12, rue de l'École-de-Médecine, 75006 Paris, France
| | - P Mordant
- Service de chirurgie thoracique, hôpital européen Georges-Pompidou, 20, rue Leblanc, 75015 Paris, France; Unversité Paris Descartes, 12, rue de l'École-de-Médecine, 75006 Paris, France
| | - M Riquet
- Service de chirurgie thoracique, hôpital européen Georges-Pompidou, 20, rue Leblanc, 75015 Paris, France; Unversité Paris Descartes, 12, rue de l'École-de-Médecine, 75006 Paris, France.
| |
Collapse
|
53
|
Zhang XY, Yin FQ, Liu L, Gao T, Ruan HY, Guan X, Lu YX, Li DR. Effects of HLEC on the secreted proteins of epithelial ovarian cancer cells prone to metastasize to lymph nodes. Cancer Biol Med 2013; 10:221-6. [PMID: 24349832 PMCID: PMC3860345 DOI: 10.7497/j.issn.2095-3941.2013.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Accepted: 09/23/2013] [Indexed: 11/23/2022] Open
Abstract
Objective To study explores the effect of HLEC on the secreted proteins of epithelial ovarian cancer (EOC) cells (SKOV3-PM4) with directional highly lymphatic metastasis. Methods Supernatants of four groups of cultured cells, namely, SKOV3 (A), SKOV3+HLEC (B), SKOV3-PM4 (C), SKOV3-PM4+HLEC (D), were collected, and their proteins were detected by antibody arrays and iTRAQ-2D-LC-MALDI-TOF/TOF/MS. Significantly differential proteins were further analyzed via bioinformatics and validated in human serums and cell media via ELISA. Results Results of antibody arrays and mass spectrometry demonstrated that GRN and VEGFA were upregulated in group C (compared with group A), whereas IGFBP7 and SPARC were downregulated in group D (compared with group C). Comprehensive bioinformatics analysis results showed that IGFBP7 and VEGFA were closely linked to each other. Further validation with serums showed statistical significance in VEGFA and IGFBP7 levels among groups of patients with ovarian cancers, benign tumors, and control groups. Two proteins were upegulated in the first group. VEGFA in the control group was downregulated. For IGFBP, upregulation in the control group and down-regulation in the first group were also observed. Conclusion The HLEC microenvironment is closely associated with directional metastasis to lymph nodes and with differential proteins including cell stromal proteins and adhesion factors. The upregulation of VEGFA and GRN and the downregulation of SPARC and IGFBP7 are closely associated with directional metastasis to lymph nodes in EOC cells.
Collapse
Affiliation(s)
- Xin-Ying Zhang
- The Cancer Hospital Affiliated to Guangxi Medical University, Nanning 530021, China
| | - Fu-Qiang Yin
- The Experiment Center of The Guangxi Medical University, Nanning 530021, China
| | - Li Liu
- The Cancer Hospital Affiliated to Guangxi Medical University, Nanning 530021, China
| | - Ting Gao
- The Cancer Hospital Affiliated to Guangxi Medical University, Nanning 530021, China
| | - He-Yun Ruan
- The Cancer Hospital Affiliated to Guangxi Medical University, Nanning 530021, China
| | - Xiao Guan
- The Cancer Hospital Affiliated to Guangxi Medical University, Nanning 530021, China
| | - Ying-Xin Lu
- The Cancer Hospital Affiliated to Guangxi Medical University, Nanning 530021, China
| | - Dan-Rong Li
- The Cancer Hospital Affiliated to Guangxi Medical University, Nanning 530021, China
| |
Collapse
|
54
|
Gromov P, Gromova I, Olsen CJ, Timmermans-Wielenga V, Talman ML, Serizawa RR, Moreira JM. Tumor interstitial fluid — A treasure trove of cancer biomarkers. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2013; 1834:2259-70. [DOI: 10.1016/j.bbapap.2013.01.013] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2012] [Revised: 01/09/2013] [Accepted: 01/14/2013] [Indexed: 12/11/2022]
|
55
|
Bendell JC, Gordon MS, Hurwitz HI, Jones SF, Mendelson DS, Blobe GC, Agarwal N, Condon CH, Wilson D, Pearsall AE, Yang Y, McClure T, Attie KM, Sherman ML, Sharma S. Safety, pharmacokinetics, pharmacodynamics, and antitumor activity of dalantercept, an activin receptor-like kinase-1 ligand trap, in patients with advanced cancer. Clin Cancer Res 2013; 20:480-9. [PMID: 24173543 DOI: 10.1158/1078-0432.ccr-13-1840] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
PURPOSE The angiogenesis inhibitor dalantercept (formerly ACE-041) is a soluble form of activin receptor-like kinase-1 (ALK1) that prevents activation of endogenous ALK1 by bone morphogenetic protein-9 (BMP9) and BMP10 and exhibits antitumor activity in preclinical models. This first-in-human study of dalantercept evaluated its safety, tolerability, pharmacokinetics, pharmacodynamics, and antitumor activity in adults with advanced solid tumors. EXPERIMENTAL DESIGN Patients in dose-escalating cohorts received dalantercept subcutaneously at one of seven dose levels (0.1-4.8 mg/kg) every 3 weeks until disease progression. Patients in an expansion cohort received dalantercept at 0.8 or 1.6 mg/kg every 3 weeks until disease progression. RESULTS In 37 patients receiving dalantercept, the most common treatment-related adverse events were peripheral edema, fatigue, and anemia. Edema and fluid retention were dose-limiting toxicities and responded to diuretic therapy. No clinically significant, treatment-related hypertension, proteinuria, gross hemorrhage, or gastrointestinal perforations were observed. One patient with refractory squamous cell cancer of the head and neck had a partial response, and 13 patients had stable disease according to RECISTv1.1, eight of whom had prolonged periods (≥12 weeks) of stable disease. Correlative pharmacodynamic markers included tumor metabolic activity and tumor blood flow, which decreased from baseline in 63% and 82% of evaluable patients, respectively, and telangiectasia in eight patients. CONCLUSION Dalantercept was well-tolerated at doses up to 1.6 mg/kg, with a safety profile distinct from inhibitors of the VEGF pathway. Dalantercept displayed promising antitumor activity in patients with advanced refractory cancer, and multiple phase II studies are underway.
Collapse
Affiliation(s)
- Johanna C Bendell
- Authors' Affiliations: Sarah Cannon Research Institute, Nashville, Tennessee; Pinnacle Oncology Hematology, Scottsdale, Arizona; Duke University Medical Center, Durham, North Carolina; Huntsman Cancer Institute, Salt Lake City, Utah; and Acceleron Pharma, Cambridge, Massachusetts
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
56
|
Jian M, Qingfu Z, Yanduo J, Guocheng J, Xueshan Q. Anti-lymphangiogenesis effects of a specific anti-interleukin 7 receptor antibody in lung cancer model in vivo. Mol Carcinog 2013; 54:148-55. [PMID: 24115038 DOI: 10.1002/mc.22082] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2013] [Revised: 07/19/2013] [Accepted: 08/07/2013] [Indexed: 12/12/2022]
Abstract
Interleukin 7 (IL-7) is known to promote lymphangiogenesis. To study the relationship between IL-7 and the lymphangiogenesis in lung cancer cells xenograft tumors, we investigated how IL-7 regulates lymphangiogenesis by Quantitative real-time reverse transcriptase-polymerase chain reaction, Western blot, co-immunoprecipitation, chromatin immunoprecipitation, and immunohistochemistry methods. We found that, in lung cancer cells xenograft tumors IL-7/IL-7 receptor (IL-7R) increase the expression of VEGF-D and lymphangiogenesis, induce c-Fos and c-Jun heterodimer formation, and enhance c-Fos/c-Jun DNA binding activity to regulate VEGF-D. Taken together, our results provided evidence that IL-7/IL-7R induce VEGF-D upregulation and promote lymphangiogenesis via c-Fos/c-Jun pathway in lung cancer.
Collapse
Affiliation(s)
- Ming Jian
- No. 202 Hospital of People Liberation Army of China, Shenyang, P.R., China
| | | | | | | | | |
Collapse
|
57
|
Inhibition of lymphangiogenesis and angiogenesis in breast tumor xenografts and lymph nodes by a peptide derived from transmembrane protein 45A. Neoplasia 2013; 15:112-24. [PMID: 23441126 DOI: 10.1593/neo.121638] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2012] [Revised: 12/11/2012] [Accepted: 12/11/2012] [Indexed: 12/17/2022] Open
Abstract
Angiogenesis, the formation of new blood vessels from preexisting blood vessels, is a process that supports tumor growth and metastatic dissemination. Lymphangiogenesis also facilitates metastasis by increasing dissemination through the lymphatic vessels (LVs). Even after treatment with antiangiogenic agents, breast cancer patients are vulnerable to LV-mediated metastasis. We report that a 14-amino acid peptide derived from transmembrane protein 45A shows multimodal inhibition of lymphangiogenesis and angiogenesis in breast cancer. The peptide blocks lymphangiogenic and angiogenic phenotypes of lymphatic and blood endothelial cells induced by tumor-conditioned media prepared from MDA-MB-231 breast cancer cells. The peptide delays growth of MDA-MB-231 tumor xenografts and normalizes tumor-conditioned lymph nodes (LNs). These studies demonstrate the antilymphangiogenic and antiangiogenic potential of the peptide against primary tumors and premetastatic, tumor-conditioned regional LNs. Mechanistically, the peptide blocks vascular endothelial growth factor receptors 2 and 3 (VEGFR2/3) and downstream proteins by binding to neuropilin 1/2 (NRP1/2) and inhibiting VEGFR2/3 and NRP1/2 complex formation in the presence of VEGFA/C.
Collapse
|
58
|
Huang YH, Yang HY, Hsu YF, Chiu PT, Ou G, Hsu MJ. Src contributes to IL6-induced vascular endothelial growth factor-C expression in lymphatic endothelial cells. Angiogenesis 2013; 17:407-18. [DOI: 10.1007/s10456-013-9386-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2012] [Accepted: 09/12/2013] [Indexed: 12/20/2022]
|
59
|
Gogineni A, Caunt M, Crow A, Lee CV, Fuh G, van Bruggen N, Ye W, Weimer RM. Inhibition of VEGF-C modulates distal lymphatic remodeling and secondary metastasis. PLoS One 2013; 8:e68755. [PMID: 23874750 PMCID: PMC3712991 DOI: 10.1371/journal.pone.0068755] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2012] [Accepted: 06/04/2013] [Indexed: 12/26/2022] Open
Abstract
Tumor-associated lymphatics are postulated to provide a transit route for disseminating metastatic cells. This notion is supported by preclinical findings that inhibition of pro-lymphangiogenic signaling during tumor development reduces cell spread to sentinel lymph nodes (SLNs). However, it is unclear how lymphatics downstream of SLNs contribute to metastatic spread into distal organs, or if modulating distal lymph transport impacts disease progression. Utilizing murine models of metastasis, longitudinal in vivo imaging of lymph transport, and function blocking antibodies against two VEGF family members, we provide evidence that distal lymphatics undergo disease course-dependent up-regulation of lymph transport coincidental with structural remodeling. Inhibition of VEGF-C activity with antibodies against VEGF-C or NRP2 prevented these disease-associated changes. Furthermore, utilizing a novel model of adjuvant treatment, we demonstrate that antagonism of VEGF-C or NRP2 decreases post SLN metastasis. These data support a potential therapeutic strategy for inhibiting distant metastatic dissemination via targeting tumor-associated lymphatic remodeling.
Collapse
Affiliation(s)
- Alvin Gogineni
- Department of Biomedical Imaging, Genentech Inc., South San Francisco, California, United States of America
| | - Maresa Caunt
- Department of Molecular Biology, Genentech Inc., South San Francisco, California, United States of America
| | - Ailey Crow
- Department of Molecular Biology, Genentech Inc., South San Francisco, California, United States of America
| | - Chingwei V. Lee
- Department of Antibody Engineering, Genentech Inc., South San Francisco, California, United States of America
| | - Germaine Fuh
- Department of Antibody Engineering, Genentech Inc., South San Francisco, California, United States of America
| | - Nicholas van Bruggen
- Department of Biomedical Imaging, Genentech Inc., South San Francisco, California, United States of America
| | - Weilan Ye
- Department of Molecular Biology, Genentech Inc., South San Francisco, California, United States of America
| | - Robby M. Weimer
- Department of Biomedical Imaging, Genentech Inc., South San Francisco, California, United States of America
- * E-mail:
| |
Collapse
|
60
|
Elsir T, Smits A, Lindström MS, Nistér M. Transcription factor PROX1: its role in development and cancer. Cancer Metastasis Rev 2013; 31:793-805. [PMID: 22733308 DOI: 10.1007/s10555-012-9390-8] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The homeobox gene PROX1 is critical for organ development during embryogenesis. The Drosophila homologue, known as prospero has been shown to act as a tumor suppressor by controlling asymmetric cell division of neuroblasts. Likewise, alterations in PROX1 expression and function are associated with a number of human cancers including hematological malignancies, carcinomas of the pancreas, liver and the biliary system, sporadic breast cancer, Kaposiform hemangioendothelioma, colon cancer, and brain tumors. PROX1 is involved in cancer development and progression and has been ascribed both tumor suppressive and oncogenic properties in a variety of different cancer types. However, the exact mechanisms through which PROX1 regulates proliferation, migration, and invasion of cancer cells are by large unknown. This review provides an update on the role of PROX1 in organ development and on its emerging functions in cancer, with special emphasis on the central nervous system and glial brain tumors.
Collapse
Affiliation(s)
- Tamador Elsir
- Department of Oncology-Pathology, Karolinska Institutet, CCK R8:05, Karolinska University Hospital, 17176 Stockholm, Sweden.
| | | | | | | |
Collapse
|
61
|
Bayer CL, Joshi PP, Emelianov SY. Photoacoustic imaging: a potential tool to detect early indicators of metastasis. Expert Rev Med Devices 2013; 10:125-34. [PMID: 23278229 DOI: 10.1586/erd.12.62] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The metastasis of cancer is a multistage process involving complex biological interactions and difficult to predict outcomes. Accurate assessment of the extent of metastasis is critical for clinical practice; unfortunately, medical imaging methods capable of identifying the early stages of invasion and metastasis are lacking. Photoacoustic imaging is capable of providing noninvasive, real-time imaging of significant anatomical and physiological changes. indicating the progression of cancer invasion and metastasis. Preclinically, photoacoustic methods have been used to image lymphatic anatomy, including the sentinel lymph nodes, to identify circulating tumor cells within vasculature and to detect micrometastases. Progress has begun toward the development of clinically applicable photoacoustic imaging systems to assist with the determination of cancer stage and likelihood of metastatic invasion.
Collapse
Affiliation(s)
- Carolyn L Bayer
- Department of Biomedical Engineering, The University of Texas at Austin, 1 University Station, Austin, TX 78712, USA.
| | | | | |
Collapse
|
62
|
Albrecht I, Bieri R, Leu A, Granacher P, Hagmann J, Kilimann MW, Christofori G. Paralemmin-1 is expressed in lymphatic endothelial cells and modulates cell migration, cell maturation and tumor lymphangiogenesis. Angiogenesis 2013; 16:795-807. [PMID: 23709172 DOI: 10.1007/s10456-013-9356-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2013] [Accepted: 05/19/2013] [Indexed: 12/16/2022]
Abstract
The lymphatic system, the network of lymphatic vessels and lymphoid organs, maintains the body fluid balance and ensures the immunological surveillance of the body. In the adult organism, the de novo formation of lymphatic vessels is mainly observed in pathological conditions. In contrast to the molecular mechanisms governing the generation of the lymphatic vasculature during embryogenesis, the processes underlying pathological lymphangiogenesis are less well understood. A genome-wide screen comparing the transcriptome of tumor-derived lymphatic endothelial cells with that of blood vessel endothelial cells identified paralemmin-1 as a protein prominently expressed in lymphatic endothelial cells. Paralemmin-1 is a lipid-anchored membrane protein that in fibroblasts and neurons plays a role in the regulation of cell shape, plasma membrane dynamics and cell motility. Here, we show that paralemmin-1 is expressed in tumor-derived lymphatic endothelial cells as well as in lymphatic endothelial cells of normal, non-tumorigenic tissue. Paralemmin-1 represses cell migration and delays the formation of tube-like structures of lymphatic endothelial cells in vitro by modulating cell-substrate adhesion, filopodia formation and plasma membrane blebbing. While constitutive genetic ablation of paralemmin-1 expression in mice has no effect on the development and physiological function of the lymphatic system, the loss of paralemmin-1 impaired tumor-associated lymphangiogenesis. Together, these results newly identify paralemmin-1 as a protein highly expressed in lymphatic endothelial cells. Similar to its function in neurons, it may link the cytoskeleton to the plasma membrane and thereby modulate lymphatic endothelial cell adhesion, migration and lymphangiogenesis.
Collapse
Affiliation(s)
- Imke Albrecht
- Department of Biomedicine, University of Basel, Mattenstrasse 28, 4058, Basel, Switzerland
| | | | | | | | | | | | | |
Collapse
|
63
|
Karnezis T, Shayan R, Fox S, Achen MG, Stacker SA. The connection between lymphangiogenic signalling and prostaglandin biology: a missing link in the metastatic pathway. Oncotarget 2012; 3:893-906. [PMID: 23097685 PMCID: PMC3478465 DOI: 10.18632/oncotarget.593] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2012] [Accepted: 08/17/2012] [Indexed: 12/21/2022] Open
Abstract
Substantial evidence supports important independent roles for lymphangiogenic growth factor signaling and prostaglandins in the metastatic spread of cancer. The significance of the lymphangiogenic growth factors, vascular endothelial growth factor (VEGF)-C and VEGF-D, is well established in animal models of metastasis, and a strong correlation exits between an increase in expression of VEGF-C and VEGF-D, and metastatic spread in various solid human cancers. Similarly, key enzymes that control the production of prostaglandins, cyclooxygenases (COX-1 and COX-2, prototypic targets of Non-steroidal anti-inflammatory drugs (NSAIDs)), are frequently over-expressed or de-regulated in the progression of cancer. Recent data have suggested an intersection of lymphangiogenic growth factor signaling and the prostaglandin pathways in the control of metastatic spread via the lymphatic vasculature. Furthermore, this correlates with current clinical data showing that some NSAIDs enhance the survival of cancer patients through reducing metastasis. Here, we discuss the potential biochemical and cellular basis for such anti-cancer effects of NSAIDs through the prostaglandin and VEGF signaling pathways.
Collapse
Affiliation(s)
- Tara Karnezis
- Peter MacCallum Cancer Centre, Locked Bag 1, A'Beckett Street, East Melbourne, Victoria, Australia
| | | | | | | | | |
Collapse
|