1
|
Zhao C, Liu Y, Meng J, Wang X, Liu X, Li W, Zhou Q, Xiang J, Li N, Hou S. LGALS3BP in Microglia Promotes Retinal Angiogenesis Through PI3K/AKT Pathway During Hypoxia. Invest Ophthalmol Vis Sci 2022; 63:25. [PMID: 35895036 PMCID: PMC9344220 DOI: 10.1167/iovs.63.8.25] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Purpose Retinal microglia promote angiogenesis and vasculopathy in oxygen-induced retinopathy (OIR); however, its specific molecular mechanism in the formation of retinal angiogenesis remains unclear. The lectin galactoside-binding soluble 3 binding protein (LGALS3BP), a member of the scavenger receptor cysteine-rich (SRCR) domain protein family, is involved in tumor neovascularization, and we therefore hypothesized that LGALS3BP plays an active role in microglia-induced angiogenesis. Methods The expression of LGALS3BP in microglia was detected by immunofluorescence, RT-qPCR, and western blotting. Functional assays of human umbilical vein endothelial cells (HUVECs) such as migration, proliferation, and tube formation were measured by Transwell, EdU, and Matrigel assays. Angiogenesis-related factors and PI3K/AKT levels were detected by western blotting. The relationship between LGALS3BP and PI3K or HIF-1α was investigated by immunoprecipitation. Results Our results showed that the expression of LGALS3BP was significantly increased in microglia surrounding neovascularization of the OIR mice and was also upregulated in human microglial clone 3 (HMC3) cells after hypoxia. Moreover, HUVECs co-cultured with hypoxic HMC3 cells showed increased migration, proliferation, and tube formation, as well as levels of angiogenesis-related factor. However, the proangiogenic ability and angiogenesis-related factor expression of HMC3 cells was suppressed after silencing LGALS3BP. LGALS3BP induces the upregulation of angiogenesis-related factors through the PI3K/AKT pathway and then promotes angiogenesis in microglia. Conclusions Collectively, our findings suggest that LGALS3BP in microglia plays an important role in angiogenesis, suggesting a potential therapeutic target of LGALS3BP for angiogenesis.
Collapse
Affiliation(s)
- Chenyang Zhao
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Ophthalmology, Chongqing, China.,Chongqing Branch of National Clinical Research Center for Ocular Diseases, Chongqing, China
| | - Yusen Liu
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Ophthalmology, Chongqing, China.,Chongqing Branch of National Clinical Research Center for Ocular Diseases, Chongqing, China
| | - Jiayu Meng
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Ophthalmology, Chongqing, China.,Chongqing Branch of National Clinical Research Center for Ocular Diseases, Chongqing, China
| | - Xiaotang Wang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Ophthalmology, Chongqing, China.,Chongqing Branch of National Clinical Research Center for Ocular Diseases, Chongqing, China
| | - Xianyang Liu
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Ophthalmology, Chongqing, China.,Chongqing Branch of National Clinical Research Center for Ocular Diseases, Chongqing, China
| | - Wanqian Li
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Ophthalmology, Chongqing, China.,Chongqing Branch of National Clinical Research Center for Ocular Diseases, Chongqing, China
| | - Qian Zhou
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Ophthalmology, Chongqing, China.,Chongqing Branch of National Clinical Research Center for Ocular Diseases, Chongqing, China
| | - Junjie Xiang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Ophthalmology, Chongqing, China.,Chongqing Branch of National Clinical Research Center for Ocular Diseases, Chongqing, China
| | - Na Li
- College of Basic Medicine, Chongqing Medical University, Chongqing, China
| | - Shengping Hou
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Ophthalmology, Chongqing, China.,Chongqing Branch of National Clinical Research Center for Ocular Diseases, Chongqing, China
| |
Collapse
|
2
|
Immunological Aspects of Age-Related Macular Degeneration. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1256:143-189. [PMID: 33848001 DOI: 10.1007/978-3-030-66014-7_6] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Increasing evidence over the past two decades points to a pivotal role for immune mechanisms in age-related macular degeneration (AMD) pathobiology. In this chapter, we will explore immunological aspects of AMD, with a specific focus on how immune mechanisms modulate clinical phenotypes of disease and severity and how components of the immune system may serve as triggers for disease progression in both dry and neovascular AMD. We will briefly review the biology of the immune system, defining the role of immune mechanisms in chronic degenerative disease and differentiating from immune responses to acute injury or infection. We will explore current understanding of the roles of innate immunity (especially macrophages), antigen-specific immunity (T cells, B cells, and autoimmunity), immune amplifications systems, especially complement activity and the NLRP3 inflammasome, in the pathogenesis of both dry and neovascular AMD, reviewing data from pathology, experimental animal models, and clinical studies of AMD patients. We will also assess how interactions between the immune system and infectious pathogens could potentially modulate AMD pathobiology via alterations in in immune effector mechanisms. We will conclude by reviewing the paradigm of "response to injury," which provides a means to integrate various immunologic mechanisms along with nonimmune mechanisms of tissue injury and repair as a model to understand the pathobiology of AMD.
Collapse
|
3
|
Tumor-Associated Macrophages and Mast Cells Positive to Tryptase Are Correlated with Angiogenesis in Surgically-Treated Gastric Cancer Patients. Int J Mol Sci 2018; 19:ijms19041176. [PMID: 29649166 PMCID: PMC5979483 DOI: 10.3390/ijms19041176] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 04/09/2018] [Accepted: 04/09/2018] [Indexed: 12/20/2022] Open
Abstract
Mast cells and macrophages can play a role in tumor angiogenesis by stimulating microvascular density (MVD). The density of mast cells positive to tryptase (MCDPT), tumor-associated macrophages (TAMs), and MVD were evaluated in a series of 86 gastric cancer (GC) tissue samples from patients who had undergone potential curative surgery. MCDPT, TAMs, and MVD were assessed in tumor tissue (TT) and in adjacent normal tissue (ANT) by immunohistochemistry and image analysis. Each of the above parameters was correlated with the others and, in particular for TT, with important clinico-pathological features. In TT, a significant correlation between MCDPT, TAMs, and MVD was found by Pearson t-test analysis (p ranged from 0.01 to 0.02). No correlation to the clinico-pathological features was found. A significant difference in terms of mean MCDPT, TAMs, and MVD between TT and ANT was found (p ranged from 0.001 to 0.002). Obtained data suggest MCDPT, TAMs, and MVD increased from ANT to TT. Interestingly, MCDPT and TAMs are linked in the tumor microenvironment and they play a role in GC angiogenesis in a synergistic manner. The assessment of the combination of MCDPT and TAMs could represent a surrogate marker of angiogenesis and could be evaluated as a target of novel anti-angiogenic therapies in GC patients.
Collapse
|
4
|
Ammendola M, Patruno R, Sacco R, Marech I, Sammarco G, Zuccalà V, Luposella M, Zizzo N, Gadaleta C, Porcelli M, Gadaleta CD, Ribatti D, Ranieri G. Mast cells positive to tryptase and tumour-associated macrophages correlate with angiogenesis in locally advanced colorectal cancer patients undergone to surgery. Expert Opin Ther Targets 2016; 20:533-40. [PMID: 26914851 DOI: 10.1517/14728222.2016.1158811] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
OBJECTIVE The density of mast cells positive to tryptase (MCDPT) and tumor-associated macrophages (TAMs) were evaluated in a series of 87 patients with stage B and C colorectal cancer who had undergone radical surgery. METHODS MCDPT, TAMs, microvascular density (MVD), endothelial area (EA) and CD8(+) tumor infiltrating lymphocytes (CD8(+) TILs) were evaluated in tumor tissue samples by immunohistochemistry and image analysis. Each of the above parameters was correlated with the others and with the main clinico-pathological features. RESULTS A significant correlation between MCDPT, TAMs, MVD and EA was found by Pearson t-test analysis. With special references to the clinico-pathological features a minimal correlation using univariate analysis was found but it was not retained at multivariate analysis. CONCLUSIONS Our data suggest that MCDPT and TAMs are linked in the tumor microenvironment and play a role in CRC angiogenesis in a synergistic manner. The assessment of the combination MCDPT and TAMs could be evaluated as a target of novel anti-angiogenic therapies in colorectal cancer patients.
Collapse
Affiliation(s)
- Michele Ammendola
- a Department of Medical and Surgery Science, Clinical Surgery Unit , University of Catanzaro 'Magna Graecia' Medical School , Catanzaro , Italy.,b Surgery Unit , National Cancer Research Centre, Giovanni Paolo II , Bari , Italy
| | - Rosa Patruno
- c Diagnostic and Interventional Radiology Unit with Integrated Section of Translational Medical Oncology , National Cancer Research Centre, 'Giovanni Paolo II' , Bari , Italy
| | - Rosario Sacco
- a Department of Medical and Surgery Science, Clinical Surgery Unit , University of Catanzaro 'Magna Graecia' Medical School , Catanzaro , Italy
| | - Ilaria Marech
- c Diagnostic and Interventional Radiology Unit with Integrated Section of Translational Medical Oncology , National Cancer Research Centre, 'Giovanni Paolo II' , Bari , Italy
| | - Giuseppe Sammarco
- a Department of Medical and Surgery Science, Clinical Surgery Unit , University of Catanzaro 'Magna Graecia' Medical School , Catanzaro , Italy
| | - Valeria Zuccalà
- d Health Science Department, Pathology Unit , University of Catanzaro 'Magna Graecia' Medical School , Catanzaro , Italy
| | - Maria Luposella
- e Department of Medical and Surgery Science, Cardiovascular Disease Unit , University of Catanzaro 'Magna Graecia' Medical School , Catanzaro , Italy
| | - Nicola Zizzo
- f Chair of Pathology, Veterinary Medical School , University of Bari , Bari , Italy
| | - Claudia Gadaleta
- c Diagnostic and Interventional Radiology Unit with Integrated Section of Translational Medical Oncology , National Cancer Research Centre, 'Giovanni Paolo II' , Bari , Italy
| | - Mariangela Porcelli
- c Diagnostic and Interventional Radiology Unit with Integrated Section of Translational Medical Oncology , National Cancer Research Centre, 'Giovanni Paolo II' , Bari , Italy
| | - Cosmo Damiano Gadaleta
- c Diagnostic and Interventional Radiology Unit with Integrated Section of Translational Medical Oncology , National Cancer Research Centre, 'Giovanni Paolo II' , Bari , Italy
| | - Domenico Ribatti
- g Department of Basic Medical Sciences, Neurosciences and Sensory Organs , University of Bari Medical School , Bari , Italy.,h National Cancer Institute 'Giovanni Paolo II' , Bari , Italy
| | - Girolamo Ranieri
- c Diagnostic and Interventional Radiology Unit with Integrated Section of Translational Medical Oncology , National Cancer Research Centre, 'Giovanni Paolo II' , Bari , Italy
| |
Collapse
|
5
|
Ammendola M, Sacco R, Sammarco G, Donato G, Montemurro S, Ruggieri E, Patruno R, Marech I, Cariello M, Vacca A, Gadaleta CD, Ranieri G. Correlation between serum tryptase, mast cells positive to tryptase and microvascular density in colo-rectal cancer patients: possible biological-clinical significance. PLoS One 2014; 9:e99512. [PMID: 24915568 PMCID: PMC4051753 DOI: 10.1371/journal.pone.0099512] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Accepted: 05/15/2014] [Indexed: 02/07/2023] Open
Abstract
Background Tryptase is a serin protease stored and released from mast cells (MCs) that plays a role in tumour angiogenesis. In this study we aimed to evaluate serum tryptase levels in colo-rectal cancer (CRC) patients before (STLBS) and after (STLAS) radical surgical resection. We also evaluated mast cell density positive to tryptase (MCDPT) and microvascular density (MVD) in primary tumour tissue. Methods A series of 61 patients with stage B and C CRC (according to the Astler and Coller staging system) were selected. Serum blood samples were collected from patients one day before and one day after surgery. Tryptase levels were measured using the UniCAP Tryptase Fluoroenzymeimmunoassay (Pharmacia, Uppsala, Sweden). Tumour sections were immunostained with a primary anti-tryptase antibody (clone AA1; Dako, Glostrup, Denmark) and an anti CD-34 antibody (QB-END 10; Bio-Optica Milan, Italy) by means of immunohistochemistry and then evaluated by image analysis methods. Results The mean ± s.d. STLBS and STLAS was 5.63±2.61 µg/L, and 3.39±1.47 µg/L respectively and a significant difference between mean levels was found: p = 0.000 by t-test. The mean ± s.d. of MCDPT and MVD was 8.13±3.28 and 29.16±7.39 respectively. A strong correlation between STLBS and MVD (r = 0.83, p = 0.000); STLBS and MCDPT (r = 0.60, p = 0.003); and MCDPT and MVD (r = 0.73; p = 0.001) was found. Conclusion Results demonstrated higher STLBS in CRC patients, indicating an involvement of MC tryptase in CRC angiogenesis. Data also indicated lower STLAS, suggesting the release of tryptase from tumour-infiltrating MCs. Serum tryptase levels may therefore play a role as a novel bio-marker predictive of response to radical surgery. In this context tryptase inhibitors such as Gabexate and Nafamostat Mesilate might be evaluated in adjuvant clinical trials as a new anti-angiogenic approach.
Collapse
Affiliation(s)
- Michele Ammendola
- Department of Medical and Surgery Science, Clinical Surgery Unit, University of Catanzaro “Magna Graecia” Medical School, Catanzaro, Italy
- Surgery Unit, National Cancer Research Centre Istituto Tumori “Giovanni Paolo II”, Bari, Italy
| | - Rosario Sacco
- Department of Medical and Surgery Science, Clinical Surgery Unit, University of Catanzaro “Magna Graecia” Medical School, Catanzaro, Italy
| | - Giuseppe Sammarco
- Department of Medical and Surgery Science, Clinical Surgery Unit, University of Catanzaro “Magna Graecia” Medical School, Catanzaro, Italy
| | - Giuseppe Donato
- Department of Medical and Surgery Science, Pathology Unit, University of Catanzaro “Magna Graecia” Medical School, Catanzaro, Italy
| | - Severino Montemurro
- Surgery Unit, National Cancer Research Centre Istituto Tumori “Giovanni Paolo II”, Bari, Italy
| | - Eustachio Ruggieri
- Surgery Unit, National Cancer Research Centre Istituto Tumori “Giovanni Paolo II”, Bari, Italy
| | | | - Ilaria Marech
- Interventional Radiology Unit with Integrated Section of Translational Medical Oncology, National Cancer Research Centre Istituto Tumori “Giovanni Paolo II”, Bari, Italy
| | - Marica Cariello
- Interventional Radiology Unit with Integrated Section of Translational Medical Oncology, National Cancer Research Centre Istituto Tumori “Giovanni Paolo II”, Bari, Italy
| | - Angelo Vacca
- Department of Biomedical Science and Human Oncology Clinica Medica “G. Baccelli”, University of Bari Medical School, Bari, Italy
| | - Cosmo Damiano Gadaleta
- Interventional Radiology Unit with Integrated Section of Translational Medical Oncology, National Cancer Research Centre Istituto Tumori “Giovanni Paolo II”, Bari, Italy
| | - Girolamo Ranieri
- Interventional Radiology Unit with Integrated Section of Translational Medical Oncology, National Cancer Research Centre Istituto Tumori “Giovanni Paolo II”, Bari, Italy
- * E-mail:
| |
Collapse
|
6
|
Sekhon BK, Sze DMY, Chan WK, Fan K, Li GQ, Moore DE, Roubin RH. PSP activates monocytes in resting human peripheral blood mononuclear cells: immunomodulatory implications for cancer treatment. Food Chem 2012; 138:2201-9. [PMID: 23497877 DOI: 10.1016/j.foodchem.2012.11.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2012] [Revised: 09/19/2012] [Accepted: 11/06/2012] [Indexed: 12/31/2022]
Abstract
Polysaccharopeptide (PSP), from Coriolus versicolor, has been used as an adjuvant to chemotherapy, and has demonstrated anti-tumor and immunomodulating effects. However its mechanism remains unknown. To elucidate how PSP affects immune populations, we compared PSP treatments both with and without prior incubation in phytohaemagglutinin (PHA) - a process commonly used in immune population experimentation. We first standardised a capillary electrophoresis fingerprinting technique for PSP identification and characterisation. We then established the proliferative capability of PSP on various immune populations in peripheral blood mononuclear cells, using flow cytometry, without prior PHA treatment. It was found that PSP significantly increased the number of monocytes (CD14(+)/CD16(-)) compared to controls without PHA. This increase in monocytes was confirmed using another antibody panel of CD14 and MHCII. In contrast, proliferations of T-cells, NK, and B-cells were not significantly changed by PSP. Thus, stimulating monocyte/macrophage function with PSP could be an effective therapeutic intervention in targeting tumors.
Collapse
|
7
|
Vascular endothelial growth factor A gene (VEGFA) polymorphisms and expression of VEGFA gene in lung cancer patients of Kashmir Valley (India). Tumour Biol 2012; 33:833-9. [DOI: 10.1007/s13277-011-0306-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2011] [Accepted: 12/21/2011] [Indexed: 01/22/2023] Open
|
8
|
Payne SJL, Jones L. Influence of the tumor microenvironment on angiogenesis. Future Oncol 2011; 7:395-408. [PMID: 21417903 DOI: 10.2217/fon.11.13] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
It is becoming increasingly recognized that the host microenvironment is essential for regulating tumor cell behavior. The cellular stromal compartment can modulate angiogenesis either directly through enhanced secretion of pro-angiogenic factors or reduced secretion of antiangiogenic factors, or indirectly by modulating the surrounding extracellular matrix. Control of angiogenesis represents a critical step in cancer progression and is a potential therapeutic target. This article focuses on the role of the tumor microenvironment in the control of angiogenesis and how dissection of the molecular interactions may enhance prognostic and predictive power and facilitate therapeutic targeting.
Collapse
Affiliation(s)
- Sarah J L Payne
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK.
| | | |
Collapse
|
9
|
Karlou M, Tzelepi V, Efstathiou E. Therapeutic targeting of the prostate cancer microenvironment. Nat Rev Urol 2011; 7:494-509. [PMID: 20818327 DOI: 10.1038/nrurol.2010.134] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Solid tumors can be thought of as multicellular 'organs' that consist of a variety of cells as well as a scaffold of noncellular matrix. Stromal-epithelial crosstalk is integral to prostate cancer progression and metastasis, and androgen signaling is an important component of this crosstalk at both the primary and metastatic sites. Intratumoral production of androgen is an important mechanism of castration resistance and has been the focus of novel therapeutic approaches with promising results. Various other pathways are important for stromal-epithelial crosstalk and represent attractive candidate therapeutic targets. Hedgehog signaling has been associated with tumor progression, growth and survival, while Src family kinases have been implicated in tumor progression and in regulation of cancer cell migration. Fibroblast growth factors and transforming growth factor beta signaling regulate cell proliferation, apoptosis and angiogenesis in the prostate cancer microenvironment. Integrins mediate communication between the cell and the extracellular matrix, enhancing growth, migration, invasion and metastasis of cancer cells. The contribution of stromal-epithelial crosstalk to prostate cancer initiation and progression provides the impetus for combinatorial microenvironment-targeting strategies.
Collapse
Affiliation(s)
- Maria Karlou
- Department of Genitourinary Medical Oncology, David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77230-1439, USA
| | | | | |
Collapse
|
10
|
Abraham D, Zins K, Sioud M, Lucas T, Schäfer R, Stanley ER, Aharinejad S. Stromal cell-derived CSF-1 blockade prolongs xenograft survival of CSF-1-negative neuroblastoma. Int J Cancer 2010; 126:1339-52. [PMID: 19711348 DOI: 10.1002/ijc.24859] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The molecular mechanisms of tumor-host interactions that render neuroblastoma (NB) cells highly invasive are unclear. Cancer cells upregulate host stromal cell colony-stimulating factor-1 (CSF-1) production to recruit tumor-associated macrophages (TAMs) and accelerate tumor growth by affecting extracellular matrix remodeling and angiogenesis. By coculturing NB with stromal cells in vitro, we showed the importance of host CSF-1 expression for macrophage recruitment to NB cells. To examine this interaction in NB in vivo, mice bearing human CSF-1-expressing SK-N-AS and CSF-1-negative SK-N-DZ NB xenografts were treated with intratumoral injections of small interfering RNAs directed against mouse CSF-1. Significant suppression of both SK-N-AS and SK-N-DZ NB growth by these treatments was associated with decreased TAM infiltration, matrix metalloprotease (MMP)-12 levels and angiogenesis compared to controls, while expression of tissue inhibitors of MMPs increased following mouse CSF-1 blockade. Furthermore, Tie-2-positive and -negative TAMs recruited by host CSF-1 were identified in NB tumor tissue by confocal microscopy and flow cytometry. However, host-CSF-1 blockade prolonged survival only in CSF-1-negative SK-N-DZ NB. These studies demonstrated that increased CSF-1 production by host cells enhances TAM recruitment and NB growth and that the CSF-1 phenotype of NB tumor cells adversely affects survival.
Collapse
Affiliation(s)
- Dietmar Abraham
- Laboratory for Cardiovascular Research, Vienna Medical University, A-1090Vienna, Austria
| | | | | | | | | | | | | |
Collapse
|
11
|
Al-Ansari S, Zeebregts CJ, Slart RH, Peppelenbosch M, Tio RA. Galectins in Atherosclerotic Disease. Trends Cardiovasc Med 2009; 19:164-9. [DOI: 10.1016/j.tcm.2009.10.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
12
|
Laurens N, Engelse MA, Jungerius C, Löwik CW, van Hinsbergh VWM, Koolwijk P. Single and combined effects of alphavbeta3- and alpha5beta1-integrins on capillary tube formation in a human fibrinous matrix. Angiogenesis 2009; 12:275-85. [PMID: 19449108 PMCID: PMC2752504 DOI: 10.1007/s10456-009-9150-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2008] [Accepted: 04/30/2009] [Indexed: 12/18/2022]
Abstract
The fibrinous exudate of a wound or tumor stroma facilitates angiogenesis. We studied the involvement of RGD-binding integrins during tube formation in human plasma-derived fibrin clots and human purified fibrin matrices. Capillary-like tube formation by human microvascular endothelial cells in a 3D plasma-derived fibrinous matrix was induced by FGF-2 and TNF-α and depended largely on cell-bound u-PA and plasmin activities. While tube formation was minimally affected by the addition of either the αvβ3-integrin inhibiting mAb LM609 or the α5-integrin inhibiting mAb IIA1, the general RGD-antagonist echistatin completely inhibited this process. Remarkably, when αvβ3- and α5β1-integrins were inhibited simultaneously, tube formation was reduced by 78%. It was accompanied by a 44% reduction of u-PA antigen accumulation and 41% less production of fibrin degradation products. αvβ5-integrin-blocking antibodies further enhanced the inhibition by mAb LM609 and mAb IIA1 to 94%, but had no effect by themselves. αv-specific cRGD only inhibited angiogenesis when α5β1-integrin was simultaneously blocked. Endostatin mimicked the effect of α5β1-integrin and inhibited tube formation only in the presence of LM609 or cRGD (73 and 80%, respectively). Comparable results were obtained when purified fibrin matrices were used instead of the plasma-derived fibrinous matrices. These data show that blocking of tube formation in a fibrinous exudate requires the simultaneous inhibition of αvβ3- and α5β1-integrins. This may bear impact on attempts to influence angiogenesis in a fibrinous environment.
Collapse
Affiliation(s)
- Nancy Laurens
- Laboratory for Physiology, Institute for Cardiovascular Research, VU University Medical Centre Amsterdam, Amsterdam, The Netherlands
| | | | | | | | | | | |
Collapse
|
13
|
Passantino L, Passantino G, Cianciotta A, Ribaud MR, Lo Presti G, Ranieri G, Perillo A. Expression of proto-oncogene C-kit and correlation with morphological evaluations in canine cutaneous mast cell tumors. Immunopharmacol Immunotoxicol 2008; 30:609-21. [PMID: 18608529 DOI: 10.1080/08923970801949265] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Canine cutaneous mast cell tumor (MCT) is very common disease in dogs, this is more aggressive than in other species. The biologic behavior of MCT is highly variable and a more accurate prognosis for these tumors needs to performed. The proto-oncogene c-kit is known to play a critical role in development and function of mast cells (MC). The aim of this study was to evaluate the expression of immunohistochemical pattern of c-kit in MCTs and to correlate these results with MC density (MCD) and intratumoral microvessel density (MVD). Our results confirm that a more aggressive biologic behavior of canine MCT is associated with the increased c-kit expression, further suggesting a new role for c-kit, as a useful marker, in diagnostic pathology and in tumor progression.
Collapse
Affiliation(s)
- Letizia Passantino
- Department of Animal Health and Welfare, Faculty of Veterinary Medicine, University of Bari, Bari, Italy.
| | | | | | | | | | | | | |
Collapse
|
14
|
Patruno R, Arpaia N, Gadaleta CD, Passantino L, Zizzo N, Misino A, Lucarelli NM, Catino A, Valerio P, Ribatti D, Ranieri G. VEGF concentration from plasma-activated platelets rich correlates with microvascular density and grading in canine mast cell tumour spontaneous model. J Cell Mol Med 2008; 13:555-61. [PMID: 18429933 PMCID: PMC3822515 DOI: 10.1111/j.1582-4934.2008.00355.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Canine cutaneous mast cell tumour (CMCT) is a common cutaneous tumour in dog, with a higher incidence than in human. CMCT is classified in three subgroups, well and intermediately differentiated (G1 and G2), corresponding to a benign disease, and poorly differentiated (G3), corresponding to a malignant disease, which metastasize to lymph nodes, liver, spleen and bone marrow. In this study, we have evaluated serum (S), platelet-poor plasma (P-PP), plasma-activated platelet rich (P-APR) and cytosol vascular endothelial growth factor (VEGF) concentrations, microvascular density (MVD) and mast cell density (MCD) in a series of 86 CMCTs and we have correlated these parameters with each other, by means of ELISA detection of VEGF and immunohistochemistry. Results show that VEGF level from cytosol P-APR and MVD were significantly higher in G3 CMCTs as compared to G1 or G2 subgroups. Moreover, a significantly strong correlation among VEGF levels from P-PAR and cytosol, MVD and MCD was found in G3 subgroup. Because VEGF levels from P-APR well correlated with MVD and malignancy grade in CMCT, we suggest that VEGF might be secreted from MCs and it may be a suitable surrogate inter-species angiogenetic markers of tumour progression in CMCT. Finally, CMCT seems to be a useful model to study the role of MCs in tumour angiogenesis and inhibition of MCs degranulation or activation might be a new anti-angiogenic strategy worthy to further investigations.
Collapse
Affiliation(s)
- R Patruno
- Department of Animal Health and Well-Being, University of Bari Veterinary Medical School, Bari, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Dirkx AEM, Oude Egbrink MGA, Wagstaff J, Griffioen AW. Monocyte/macrophage infiltration in tumors: modulators of angiogenesis. J Leukoc Biol 2006; 80:1183-96. [PMID: 16997855 DOI: 10.1189/jlb.0905495] [Citation(s) in RCA: 256] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The role of a tumor immune infiltrate in cancer progression and metastasis has been debated frequently. Although often considered to be associated with improved prognosis and leading to the enhanced survival of cancer patients, inflammatory cells have also been described to assist the tumor's capabilities to progress, proliferate, and metastasize. Tumor-associated macrophages (TAMs), for example, have been shown to be symbiotically related to tumor cells: Tumor cells recruit TAMs and provide them with survival factors, and TAMs in turn produce a variety of angiogenic factors in response to the tumor microenvironment. This review will describe the composition of an immune infiltrate in tumors and the angiogenic and angiostatic properties of the cells present. Special emphasis will be on the angiogenesis-associated activities of TAMs. The development of immunotherapy and gene therapy using TAMs to mediate tumor cytotoxicity or to deliver gene constructs will be discussed as well. As immunotherapy has so far not been as effective as anticipated, a combination therapy in which angiostatic agents are used as well is put forward as a novel strategy to treat cancer.
Collapse
Affiliation(s)
- Anita E M Dirkx
- Department of Pathology, University Hospital Maastricht, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands
| | | | | | | |
Collapse
|
16
|
Ranieri G, Patruno R, Lionetti A, Di Summa A, Mattioli E, Bufo P, Pellecchia A, Ribatti D, Zizzo N. Endothelial area and microvascular density in a canine non-Hodgkin's lymphoma: an interspecies model of tumor angiogenesis. Leuk Lymphoma 2006; 46:1639-43. [PMID: 16236617 DOI: 10.1080/10428190500205150] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Experimental and clinical data indicate that tumor progression is associated with angiogenesis and that an increase in microvascular density (MVD) is associated with a poor prognosis, in both solid and hematological malignancies. No data have been published concerning the relationship between angiogenesis and malignancy grade in canine non-Hodgkin's lymphoma (NHL), which is a neoplasm that shares several biological and clinical characteristics with human NHL. In the present study, we evaluated this relationship in a series of 43 cases of canine NHL. The results demonstrate that both MVD and endothelial area (EA) were significantly higher in high-grade compared to low-grade lymphoma and a good statistical correlation was found between MVD and EA. These data indicate that increased angiogenesis paralleled with increased malignancy grade in canine NHL, which represents an interesting tumor model for studying the role of angiogenesis as an interspecies pathway of tumoral malignancy and biological aggressiveness.
Collapse
Affiliation(s)
- Girolamo Ranieri
- Department of Experimental Oncology, University of Bari Medical School, Bari Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Chan JK, Magistris A, Loizzi V, Lin F, Rutgers J, Osann K, DiSaia PJ, Samoszuk M. Mast cell density, angiogenesis, blood clotting, and prognosis in women with advanced ovarian cancer. Gynecol Oncol 2005; 99:20-5. [PMID: 16055178 DOI: 10.1016/j.ygyno.2005.05.042] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2005] [Revised: 04/23/2005] [Accepted: 05/02/2005] [Indexed: 11/25/2022]
Abstract
OBJECTIVE To determine clinical or biological associations between mast cell density, blood clotting, angiogenesis, and survival of patients with advanced ovarian cancer. METHODS Tumor tissue sections were assessed for mast cell density by staining for mast cell tryptase, blood clotting by staining of thrombosed blood vessels, and angiogenesis by CD34 expression. Chi-square, Kaplan-Meier, and Cox proportional hazard statistical analyses were used. RESULTS 44 women with stage III-IV ovarian cancers had tumor blocks available for immunohistochemical analysis. Higher mean vessel density (MVD) (>11 vessels/400x field) predicted for better survival than lower MVD (< or =11 vessels/400x field) (P = 0.004). Women whose tumors had low levels of peri-tumoral mast cell infiltration had a mean survival of 40.6 months compared to 50.6 months in those whose tumors had high levels (P = 0.47). Tumors with higher MVD and high peri-tumoral mast cell infiltration had a mean survival of 80.3 months compared to 37.8 months in those with low mast cell density or low MVD (P = 0.015). Patients with tumors showing a low degree of blood clotting had a mean survival of 45.5 compared to 45.1 months in those with tumors showing a high degree of blood clotting (P = 0.91). There was no significant association between angiogenesis and mast cell density (P = 0.123). In multivariate analysis, higher MVD remained as a significant prognostic factor for improved survival after adjusting for clotting and mast cell density. CONCLUSIONS Our data suggest that peri-tumoral mast cell infiltration in tumors with high MVD predicts for improved survival in women with advanced epithelial ovarian cancer.
Collapse
Affiliation(s)
- John K Chan
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Stanford University School of Medicine, 300 Pasteur Dr. HH333, Stanford, CA 94305-5317, USA.
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Kidd KR, Dal Ponte D, Stone AL, Hoying JB, Nagle RB, Williams SK. Stimulated endothelial cell adhesion and angiogenesis with laminin-5 modification of expanded polytetrafluoroethylene. TISSUE ENGINEERING 2005; 11:1379-91. [PMID: 16259593 DOI: 10.1089/ten.2005.11.1379] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Biomedical implants often exhibit poor clinical performance due to the formation of a periimplant avascular fibrous capsule. Surface modification of synthetic materials has been evaluated to accelerate the formation of functional microcirculation in association with implants. The current study used a flow-mediated protein deposition system to modify expanded polytetrafluoroethylene (ePTFE) with a laminin-5-rich conditioned growth medium and with medium from which laminin-5 had been selectively removed. An in vitro model of endothelial cell adherence determined that laminin-5 modification resulted in significantly increased adhesion of human microvessel endothelial cells to ePTFE. In vivo studies evaluating the periimplant vascular response to laminin-5-treated samples indicated that absorption of laminin-5-rich conditioned medium supported accelerated neovascularization of ePTFE implants. A flow system designed to treat porous implant materials facilitates laminin-5 modification of commercially available ePTFE, resulting in increased endothelial cell adhesion in vitro and increased vascularization in vivo.
Collapse
Affiliation(s)
- Kameha R Kidd
- Biomedical Engineering Program University of Arizona, Tucson, Arizona 85724, USA
| | | | | | | | | | | |
Collapse
|
19
|
Kwon GY, Lee SD, Park ES. Mast cell and macrophage counts and microvessel density in invasive breast carcinoma-comparison analysis with clinicopathological parameters. Cancer Res Treat 2005; 37:103-8. [PMID: 19956488 DOI: 10.4143/crt.2005.37.2.103] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2005] [Accepted: 04/14/2005] [Indexed: 11/21/2022] Open
Abstract
PURPOSE The purpose of this study was to evaluate the clinicopathological significance of the microvessel density and macrophage and mast cell counts in invasive breast carcinomas. MATERIALS AND METHODS 45 invasive breast carcinomas were immunohistochemically stained with the endothelial antigen, CD34, and macrophage marker, CD68. 0.1% toluidine blue was used to highlight mast cells. The microvessel and mast cell counts were performed at x200 magnification and the macrophages at x400 magnification. RESULTS With the 45 invasive breast carcinomas, there were no statistically significant associations between the mast cell, macrophage and microvessel counts and the tumor size and lymph node status. ER and PR negative mast cells infiltrated more than in cases of positive stati, with statistical significance (p-value=0.010 and 0.005, respectively). The macrophage counts were negatively correlated with the PR status (p-value=0.030). With respect to the c-erbB-2 status, there was no significance correlation with the mast cell, macrophage and microvessel counts. The mast cell counts showed significantly positive correlation with the microvessel counts in the invasive breast carcinomas (p-value=0.015). In a comparison of the macrophage counts with the microvessel counts, a positive tendency for both parameters, but without statistical significance (p-value=0.310). CONCLUSION Increasing numbers of mast cells and macrophages were recruited in invasive breast carcinomas, which contribute to angiogenesis. The microvessel density in invasive breast carcinomas had no statistically significant association with the tumor size, lymph node status, and histological grade, presence of DCIS component, estrogen/progesterone receptor status and cerbB-2 status. The evaluation of angiogenesis using these methods is not thought to provide an independent clinicopathological factor in invasive breast carcinomas.
Collapse
Affiliation(s)
- Gui Young Kwon
- Department of Pathology, Chung-Ang University College of Medicine, Seoul, Korea
| | | | | |
Collapse
|
20
|
Mantzaris NV, Webb S, Othmer HG. Mathematical modeling of tumor-induced angiogenesis. J Math Biol 2004; 49:111-87. [PMID: 15293017 DOI: 10.1007/s00285-003-0262-2] [Citation(s) in RCA: 153] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2003] [Indexed: 01/06/2023]
Affiliation(s)
- Nikos V Mantzaris
- School of Mathematics, University of Minnesota, Minneapolis, MN 55455, USA
| | | | | |
Collapse
|
21
|
Schultz RM. Potential of p38 MAP kinase inhibitors in the treatment of cancer. PROGRESS IN DRUG RESEARCH. FORTSCHRITTE DER ARZNEIMITTELFORSCHUNG. PROGRES DES RECHERCHES PHARMACEUTIQUES 2003; 60:59-92. [PMID: 12790339 DOI: 10.1007/978-3-0348-8012-1_2] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
The involvement of chronic inflammation in tumor development and progression is reviewed. Based on the natural history of certain diseases and epidemiology studies, a strong association has been established between particular chronic inflammatory conditions and eventual tumor appearance. Solid tumors require a stroma for their growth and recruit macrophages to synthesize essential growth and angiogenic factors that they do not have the capacity to produce. The microenvironment of the local host tissue appears to be an active participant in exchanging cytokines and enzymes with tumor cells that modify the local extracellular matrix, stimulate migration, and promote tumor angiogenesis, proliferation and survival. The role of p38 MAP kinase as a therapeutic target for treating cancer is discussed.
Collapse
Affiliation(s)
- Richard M Schultz
- Lilly Research Laboratories, Division of Cancer Research, Indianapolis, IN 46285, USA.
| |
Collapse
|
22
|
Lockhart AC, Braun RD, Yu D, Ross JR, Dewhirst MW, Klitzman B, Yuan F, Grichnik JM, Proia AD, Conway DA, Mann G, Hurwitz HI. A clinical model of dermal wound angiogenesis. Wound Repair Regen 2003; 11:306-13. [PMID: 12846919 DOI: 10.1046/j.1524-475x.2003.11411.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Full-thickness dermal biopsies were performed in healthy volunteers to establish the range of angiogenic responses in wound healing in a normal population. Four-millimeter punch biopsies were made in the forearms of 15 healthy volunteers. Each wound was evaluated microscopically 4-5 times per week for 2 weeks. A semiquantitative wound scoring system to evaluate the neovasculature at the wound periphery was investigated. A vascular score was calculated for each wound at each observation. Two independent observers analyzed the microscopic wound images using the scoring system. At the end of the 14-day period, repeat biopsies were performed on some of the volunteers, and the granulation tissue was stained with anti-CD31. The Kaplan-Meier method was used to estimate the distribution of the time to reach predetermined target average vascular scores. A mixed-effects regression model indicated that time, age, and observer were predictors for the average vascular score outcome. The pattern and time course for wound neovascularization was highly reproducible in this group of healthy volunteers, and the assay was feasible and well tolerated. This wound angiogenesis model may be useful for monitoring the effects of antiangiogenic agents on normal wound neovascularization.
Collapse
Affiliation(s)
- A Craig Lockhart
- Department of Medicine, Duke University Medical Center, Durham, North Carolina 27710, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Lioté F, Champy R, Moenner M, Boval-Boizard B, Badet J. Elevated angiogenin levels in synovial fluid from patients with inflammatory arthritis and secretion of angiogenin by cultured synovial fibroblasts. Clin Exp Immunol 2003; 132:163-8. [PMID: 12653852 PMCID: PMC1808677 DOI: 10.1046/j.1365-2249.2003.02117.x] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Angiogenesis is a key process in the pathogenesis of inflammatory arthritis. Angiogenin is one of the most potent inducers of neovascularization in experimental models in vivo. To look for evidence that angiogenin is involved in inflammatory joint disease, we examined plasma and synovial fluid (SF) samples from rheumatology patients and synovial fibroblast cell culture supernatants. Angiogenin levels were determined by radioimmunoassay and ELISA. Plasma angiogenin concentrations ranged from 96 to 478 ng/ml, with no significant difference between patients and normal controls. In SF, angiogenin concentrations were significantly higher in patients with acute or chronic synovitis (rheumatoid arthritis (RA): median, 104 ng/ml; range 13-748, n = 14; crystal-induced arthritis (CIA): median, 149 ng/ml; range, 37-616, n = 14, and other chronic inflammatory arthritis: median, 42 ng/ml; range, 15-205; n = 9) than in the 18 patients with osteoarthritis (OA) (median, 20 ng/ml; range 8-116) (P < 0.0001, anova). Angiogenin levels in SF from RA patients in remission with secondary OA were similar to those achieved in primary OA, and decreased in parallel with the resolution of acute gout. Angiogenin protein was released by cultured synovial fibroblasts from OA and RA patients, and reached 1.18 ng/106 cells/day. These data suggest that angiogenin may mediate local inflammation in arthritis via effects on angiogenesis and leucocyte regulation.
Collapse
MESH Headings
- Analysis of Variance
- Arthritis/metabolism
- Arthritis/pathology
- Arthritis, Infectious/metabolism
- Arthritis, Infectious/pathology
- Arthritis, Psoriatic/metabolism
- Arthritis, Psoriatic/pathology
- Arthritis, Rheumatoid/metabolism
- Arthritis, Rheumatoid/pathology
- Case-Control Studies
- Cells, Cultured
- Culture Media, Conditioned/chemistry
- Fibroblasts/metabolism
- Humans
- Leukocytes, Mononuclear/chemistry
- Leukocytes, Mononuclear/metabolism
- Osteoarthritis/metabolism
- Osteoarthritis/pathology
- Ribonuclease, Pancreatic/analysis
- Ribonuclease, Pancreatic/blood
- Ribonuclease, Pancreatic/genetics
- Statistics, Nonparametric
- Synovial Fluid/chemistry
- Synovial Fluid/cytology
Collapse
Affiliation(s)
- F Lioté
- Centre Viggo Petersen, Hôpital Lariboisière, Paris, France.
| | | | | | | | | |
Collapse
|
24
|
Komorowski J, Pasieka Z, Jankiewicz-Wika J, Stepień H. Matrix metalloproteinases, tissue inhibitors of matrix metalloproteinases and angiogenic cytokines in peripheral blood of patients with thyroid cancer. Thyroid 2002; 12:655-62. [PMID: 12225633 DOI: 10.1089/105072502760258622] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Stimulation of growth of endothelial cells from preexisting blood vessels, i.e., angiogenesis, is one of the essential elements necessary to create a permissive environment in which a tumor can grow. During angiogenesis, the matrix metalloproteinase (MMP) family of tissue enzymes contributes to normal (embriogenesis or wound repair) and pathologic tissue remodeling (chronic inflammation and tumor genesis). The proposed pathogenic roles of MMPs in cancer are tissue breakdown and remodeling during invasive tumor growth and tumor angiogenesis. Tissue inhibitors of metalloproteinases (TIMPs) form a complex with MMPs, which in turn inhibits active MMPs. Vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF) are unique among mediators of angiogenesis with synergistic effect, and both can also be secreted by thyroid cancer cells. The goal of the study was to evaluate the plasma blood concentration of VEGF, bFGF, MMP-1, MMP-2, MMP-3, MMP-8, MMP-9, TIMP-1, and TIMP-2 in patients with cancer and in normal subjects. Twenty-two patients with thyroid cancers (papillary cancer, 11; partly papillary and partly follicular cancer, 3; anaplastic cancer, 5; medullary cancer, 3) and 16 healthy subjects (controls) were included in the study. VEGF, bFGF MMPs, and TIMPs were evaluated by enzyme-linked immunosorbent assay (ELISA). In patients with thyroid cancer, normal VEGF concentrations (74.29 +/- 13.38 vs. 84.85 +/- 21.71 pg/mL; p > 0.05) and increased bFGF (29.52 +/- 4.99 vs. 6.05 +/- 1.43 pg/mL; p < 0.001), MMP-2 (605.95 +/- 81.83 vs. 148.75 +/- 43.53 ng/mL; p < 0.001), TIMP-2 (114.19 +/- 6.62 vs. 60.75 +/- 9.18 ng/mL; p < 0.001), as well as lower MMP-1 (0.70 +/- 0.42 vs. 3.87 +/- 0.53; p < 0.001) levels have been noted. Increased plasma levels of MMP-3 and MMP-9 were also found in patients with medullary carcinoma. In conclusion, predominance of MMP-2 over TIMP-2 and TIMP-1 over MMP-1 as well as increased concentration of bFGF in peripheral blood are common features in patients with thyroid cancer.
Collapse
Affiliation(s)
- Jan Komorowski
- Institute of Endocrinology, Medical University of Łódź, Łódź, Poland.
| | | | | | | |
Collapse
|
25
|
Nakamura M, Abe Y, Tokunaga T. Pathological significance of vascular endothelial growth factor A isoform expression in human cancer. Pathol Int 2002; 52:331-9. [PMID: 12100515 DOI: 10.1046/j.1440-1827.2002.01367.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Vascular endothelial growth factor (VEGF) is a highly specific factor for vascular endothelial cells. Five VEGF-A isoforms (splice variants 121, 145, 165, 189 and 206) are generated as a result of alternative splicing from a single VEGF-A gene. These differ in their molecular weights and in biological properties such as their ability to bind to cell-surface heparan sulfate proteoglycans. Deregulated VEGF-A expression contributes to the development of solid tumors by promoting tumor angiogenesis. More specifically, VEGF-A189 expression is related to angiogenesis and prognosis in certain human solid tumors. VEGF-A189 expression is also related to the xenotransplantability of human cancers into immunodeficient mice in vivo. Consequently, inhibition of VEGF-A or VEGF-A189 signaling regulates the development and metastasis of a variety of tumors. This review focuses on recent studies of the mechanisms by which VEGF-A regulates angiogenesis in the cancer stroma and on our recent findings concerning the potential mechanisms of VEGF-A189 expression on tumor growth and metastasis.
Collapse
Affiliation(s)
- Masato Nakamura
- Department of Pathology, Tokai University School of Medicine, Bohseidai, Isehara, Kanagawa, Japan.
| | | | | |
Collapse
|
26
|
Jung YD, Ahmad SA, Liu W, Reinmuth N, Parikh A, Stoeltzing O, Fan F, Ellis LM. The role of the microenvironment and intercellular cross-talk in tumor angiogenesis. Semin Cancer Biol 2002; 12:105-12. [PMID: 12027582 DOI: 10.1006/scbi.2001.0418] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The expression of angiogenic factors in tumors is controlled by intrinsic factors in the tumor and by the host microenvironment. Endothelial cells within particular tumor microenvironments interact not only with tumor cells but also with immune cells, fibroblasts, pericytes, and the extracellular matrix (ECM). The relationship and cross-talk among these cells determine gene expression, phenotypic distinction, and ultimately whether endothelial cells survive, proliferate, or undergo apoptosis. The diversity of angiogenic factor expression in tumors at different sites, combined with the fact that endothelial cells in different organs and tumors are phenotypically distinct, constitutes a formidable challenge for the development of effective anti-angiogenic therapies.
Collapse
Affiliation(s)
- Young D Jung
- Department of Cancer Biology, The University of Texas, M. D. Anderson Cancer Center, Houston, Texas 77030, USA
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Rosen LS. Clinical experience with angiogenesis signaling inhibitors: focus on vascular endothelial growth factor (VEGF) blockers. Cancer Control 2002; 9:36-44. [PMID: 11965229 DOI: 10.1177/107327480200902s05] [Citation(s) in RCA: 190] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Angiogenesis is required for tumor growth and metastasis and, therefore, represents an exciting target for cancer treatment. Angiogenesis is a complex process that is tightly regulated by pro- and anti-angiogenic growth factors. Physiologic angiogenesis takes place during tissue growth and repair, during the female reproductive cycle, and during fetal development. Pathologic angiogenesis is characterized by either excessive (eg, cancer) or inadequate (eg, coronary artery disease) neovascularization. Angiogenesis occurs in a series of complex and interrelated steps that involve the release of pro-angiogenic growth factors, such as vascular endothelial growth factor (VEGF). VEGF regulates both vascular proliferation and permeability, and functions as an anti-apoptotic factor for newly formed blood vessels. The biological effects of VEGF are mediated by two receptors, VEGF-1 and VEGF-2, whose expression is largely limited to the vascular endothelium. VEGF is often expressed in tumors at substantially increased levels. It is expressed in response to hypoxia, oncogenes, and other cytokines, and its expression is associated with poor prognosis in several types of cancer. Several different strategies have been used to inhibit VEGF, including anti-VEGF monoclonal antibodies (eg, bevacizumab) and agents that inhibit the VEGF receptor (eg, SU5416). Both types of agents have tolerable side effects and have shown promise when evaluated in a wide range of tumor types. Angiogenesis, the role of VEGF in angiogenesis and malignancy, and strategies for cancer treatment with VEGF inhibitors are discussed.
Collapse
Affiliation(s)
- Lee S Rosen
- UCLA Jonsson Cancer Center, Los Angeles, CA 90095-7059, USA.
| |
Collapse
|
28
|
Kidd KR, Nagle RB, Williams SK. Angiogenesis and neovascularization associated with extracellular matrix-modified porous implants. JOURNAL OF BIOMEDICAL MATERIALS RESEARCH 2002; 59:366-77. [PMID: 11745574 DOI: 10.1002/jbm.1253] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Therapies directed toward stimulation of angiogenesis seek to accelerate the development of new blood vessels in tissues rendered dysfunctional because of an insufficient microvascular supply. The goal of the current study was the stimulation of an angiogenic response around and within porous biomedical implants, such as vascular grafts, constructed from a base polymer composed of expanded polytetrafluoroethylene (ePTFE). Similar to many biomaterials, ePTFE does not elicit a significant angiogenic response and the porous interstices of this material remain avascular after implantation. Studies were performed to evaluate the ability of a tumorigenic cell line, the 804-G rat kidney cell to secrete an angiogenic extracellular matrix on and within the porous structures of ePTFE. A rat model was used to evaluate and compare implant-associated healing responses between nonmodified materials and extracellular matrix-modified ePTFE. Results demonstrated that, in contrast to untreated ePTFE, the matrix-modified ePTFE stimulated both angiogenesis in implant-associated tissue and neovascularization of the pores within the ePTFE interstices. Deposition of an insoluble matrix stimulates an angiogenic response and has a potential application for the improvement of medical device function.
Collapse
Affiliation(s)
- Kameha R Kidd
- Physiological Sciences Graduate Program, University of Arizona, Tucson, Arizona 85724, USA
| | | | | |
Collapse
|
29
|
Ribatti D, Vacca A, Nico B, Crivellato E, Roncali L, Dammacco F. The role of mast cells in tumour angiogenesis. Br J Haematol 2001; 115:514-21. [PMID: 11736931 DOI: 10.1046/j.1365-2141.2001.03202.x] [Citation(s) in RCA: 110] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Affiliation(s)
- D Ribatti
- Department of Human Anatomy and Histology, University School, Bari, Italy.
| | | | | | | | | | | |
Collapse
|
30
|
White ES, Livant DL, Markwart S, Arenberg DA. Monocyte-fibronectin interactions, via alpha(5)beta(1) integrin, induce expression of CXC chemokine-dependent angiogenic activity. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 167:5362-6. [PMID: 11673553 DOI: 10.4049/jimmunol.167.9.5362] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Monocyte-derived macrophages are important sources of angiogenic factors in cancer and other disease states. Upon extravasation from vasculature, monocytes encounter the extracellular matrix. We hypothesized that interaction with extracellular matrix proteins leads monocytes to adopt an angiogenic phenotype. We performed endothelial cell chemotaxis assays on conditioned medium (CM) from monocytes that had been cultured in vitro on various matrix substrates (collagen I, laminin, Matrigel, fibronectin), in the presence of autologous serum, or on tissue culture plastic alone. Monocytes cultured on Matrigel and on fibronectin were the most potent inducers of angiogenic activity compared with tissue culture plastic or autologous serum-differentiated monocytes. This increased angiogenic activity was associated with increased expression of angiogenic CXC chemokines (IL-8, epithelial neutrophil-activating peptide-78, growth-related oncogene alpha, and growth-related oncogene gamma) but not of vascular endothelial growth factor. Additionally, CM from monocytes cultured on fibronectin-depleted Matrigel (MG(FN-)) induced significantly less angiogenic activity than CM from monocytes cultured on control-depleted Matrigel. ELISA analysis of CM from monocytes cultured on MG(FN-) revealed a significant decrease in GRO-alpha and GRO-gamma compared with CM from monocytes cultured on MG. Incubation of monocytes before adherence on fibronectin with PHSCN (a competitive peptide inhibitor of the PHSRN sequence of fibronectin binding via alpha(5)beta(1) integrin) results in diminished expression of angiogenic activity and CXC chemokines compared with control peptide. These data suggest that fibronectin, via alpha(5)beta(1) integrin, promotes CXC chemokine-dependent angiogenic activity from monocytes.
Collapse
Affiliation(s)
- E S White
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | | | | | | |
Collapse
|
31
|
Fujioka S, Yoshida K, Yanagisawa S, Kawakami M, Aoki T, Yamazaki Y. Angiogenesis in pancreatic carcinoma: thymidine phosphorylase expression in stromal cells and intratumoral microvessel density as independent predictors of overall and relapse-free survival. Cancer 2001; 92:1788-97. [PMID: 11745251 DOI: 10.1002/1097-0142(20011001)92:7<1788::aid-cncr1695>3.0.co;2-z] [Citation(s) in RCA: 77] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
BACKGROUND Recently, the usefulness of intratumoral microvessel density (IMD) and expression of several angiogenic factors as prognostic indicators have been demonstrated in several human solid tumors. METHODS One hundred four patients with pancreatic ductal adenocarcinoma were examined retrospectively. The investigated clinicopathologic and immunohistologic data included staining for vascular endothelial growth factor (VEGF), thymidine phosphorylase (TP), basic fibroblast growth factor (bFGF), CD34 (for calculating IMD), p53, and Ki-67. RESULTS Multivariate analysis for both overall and relapse-free survival revealed two independent variables, IMD and TP staining in stromal cells (TPs, P < 0.02). Whereas the frequency of hepatic metastasis was correlated significantly with cytoplasmic expression of TP or bFGF in tumor cells (TPc, bFGFc), IMD, and p53 status, local recurrence was significantly more common in patients with positive staining for TPs, bFGF in stromal cells (bFGFs), and for the pM category (P < 0.05). TPc, bFGFc, VEGF, and p53 expression correlated with IMD (P < 0.01), although TPs and bFGFs expression did not. VEGF and IMD status correlated with p53 expression (P < 0.001), although TP, bFGF, and Ki-67 status did not. CONCLUSIONS TPs expression and IMD were revealed to be valuable tools for predicting overall and relapse-free survival in patients with pancreatic adenocarcinoma. Whereas TPc and bFGFc are likely to participate in hepatic metastasis by means of their angiogenic properties, TPs and bFGFs may be related to local tumor progression. Angiogenesis in human pancreatic carcinoma may be dependent on VEGF, TP, and bFGF. p53 abnormality is likely to take part in VEGF-related angiogenesis.
Collapse
Affiliation(s)
- S Fujioka
- Department of Surgery 1, University Hospital, The Jikei University School of Medicine, 3-25-8, Nishi-shinbashi, Minato-ku, Tokyo 105-8461, Japan.
| | | | | | | | | | | |
Collapse
|
32
|
Ambartsumian N, Klingelhöfer J, Grigorian M, Christensen C, Kriajevska M, Tulchinsky E, Georgiev G, Berezin V, Bock E, Rygaard J, Cao R, Cao Y, Lukanidin E. The metastasis-associated Mts1(S100A4) protein could act as an angiogenic factor. Oncogene 2001; 20:4685-95. [PMID: 11498791 DOI: 10.1038/sj.onc.1204636] [Citation(s) in RCA: 182] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2000] [Revised: 04/17/2001] [Accepted: 05/09/2001] [Indexed: 12/31/2022]
Abstract
The involvement of Mts1(S100A4), a small Ca(2+)-binding protein in tumor progression and metastasis had been demonstrated. However, the mechanism by which mts1(S100A4) promoted metastasis had not been identified. Here we demonstrated that Mts1(S100A4) had significant stimulatory effect on the angiogenesis. We detected high incidence of hemangiomas--benign tumors of vascular origin in aged transgenic mice ubiquitously expressing the mts1(S100A4) gene. Furthermore, the serum level of the Mts1(S100A4) protein increased with ageing. Tumors developed in Mts1-transgenic mice revealed an enhanced vascular density. We showed that an oligomeric, but not a dimeric form of the Mts1(S100A4) protein was capable of enhancing the endothelial cell motility in vitro and stimulate the corneal neovascularization in vivo. An oligomeric fraction of the protein was detected in the conditioned media as well as in human serum. The data obtained allowed us to conclude that mts1(S100A4) might induce tumor progression via stimulation of angiogenesis.
Collapse
Affiliation(s)
- N Ambartsumian
- Department of Molecular Cancer Biology, Danish Cancer Society, DK-2100 Copenhagen Ø, Denmark.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Affiliation(s)
- A Kurup
- Department of Chemistry, Pomona College, Claremont, California 91711, USA
| | | | | |
Collapse
|
34
|
White ES, Strom SR, Wys NL, Arenberg DA. Non-small cell lung cancer cells induce monocytes to increase expression of angiogenic activity. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 166:7549-55. [PMID: 11390510 DOI: 10.4049/jimmunol.166.12.7549] [Citation(s) in RCA: 78] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Tumors are dependent on angiogenesis for survival and propagation. Accumulated evidence suggests that macrophages are a potentially important source of angiogenic factors in many disease states. However, the role(s) of macrophages in non-small cell lung cancer (NSCLC) have not been determined. We hypothesized that monocyte-derived macrophages are induced by NSCLC to increase expression of angiogenic factors. To define the role of macrophage-tumor cell interaction with respect to angiogenesis, human peripheral blood monocytes (PBM) were cocultured with A549 (human bronchoalveolar cell carcinoma) or Calu 6 (human anaplastic carcinoma) NSCLC cells. The resultant conditioned medium (CM) was evaluated for angiogenic potential and for expression of angiogenic factors. We found that endothelial cell chemotactic activity (as a measure of angiogenic potential) was significantly increased in response to CM from cocultures of PBM/NSCLC compared with PBM alone, NSCLC alone, or a combination of NSCLC and PBM CM generated separately. Subsequent analysis by ELISA reveals markedly increased CXC chemokine expression, with a lesser increase in vascular endothelial growth factor, in CM from PBM/NSCLC coculture. Neutralizing Ab to angiogenic CXC chemokines blocked the increase in endothelial cell chemotaxis. Furthermore, with separately generated CM as a stimulus, we found that macrophages are the predominant source of increased CXC chemokine expression. Finally, we found that NSCLC-derived macrophage migration-inhibitory factor is responsible for the increased expression of macrophage-derived angiogenic activity. These data suggest that the interaction between host macrophages and NSCLC cells synergistically increases angiogenic potential, and that this is due to an increased elaboration of angiogenic CXC chemokines.
Collapse
MESH Headings
- Angiogenesis Inducing Agents/biosynthesis
- Angiogenesis Inducing Agents/blood
- Carcinoma, Non-Small-Cell Lung/blood supply
- Carcinoma, Non-Small-Cell Lung/metabolism
- Carcinoma, Non-Small-Cell Lung/pathology
- Cell Line
- Cells, Cultured
- Chemokines, CXC/biosynthesis
- Chemokines, CXC/blood
- Chemotaxis, Leukocyte/physiology
- Coculture Techniques
- Culture Media, Conditioned/chemistry
- Endothelium, Vascular/cytology
- Endothelium, Vascular/physiology
- Humans
- Lung Neoplasms/blood supply
- Lung Neoplasms/metabolism
- Lung Neoplasms/pathology
- Macrophage Migration-Inhibitory Factors/biosynthesis
- Macrophage Migration-Inhibitory Factors/pharmacology
- Macrophage Migration-Inhibitory Factors/physiology
- Monocytes/metabolism
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- E S White
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | | | | | | |
Collapse
|
35
|
Bando H, Toi M. Tumor angiogenesis, macrophages, and cytokines. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2001; 476:267-84. [PMID: 10949671 DOI: 10.1007/978-1-4615-4221-6_21] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- H Bando
- Tokyo Metropolitan Komagome Hospital, Japan
| | | |
Collapse
|
36
|
Bornstein P, Kyriakides TR, Yang Z, Armstrong LC, Birk DE. Thrombospondin 2 modulates collagen fibrillogenesis and angiogenesis. J Investig Dermatol Symp Proc 2000; 5:61-6. [PMID: 11147677 DOI: 10.1046/j.1087-0024.2000.00005.x] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Thrombospondin 2 (TSP2)-null mice, generated by targeted disruption of the Thbs2 gene, display a complex phenotype that is characterized, in part, by a variety of connective tissue abnormalities and increased vascular density in skin and subcutaneous tissues. In this paper we summarize the evidence that TSP2 functions as a matricellular protein to influence cell function by modulating cell-matrix interactions, rather than acting as an integral component of the matrix. Thus, the structurally abnormal collagen fibrils detected in skin appear to be the consequence of the defective adhesion demonstrated by dermal fibroblasts in culture that, in turn, result from increased matrix metalloproteinase 2 (MMP2, gelatinase A) production by these cells. Corroborating evidence for such a mode of action comes from transmission electron microscopic images of developing flexor muscle tendons that show distinct abnormalities in fibroblast-collagen fibril interactions in TSP2-null tissue. The increased vascular density seen in skin of TSP2-null mice can be reproduced in a number of models of injury, including subcutaneous implantation of polyvinyl alcohol sponges and silicone rubber discs, and excisional skin wounds. Experiments are proposed to distinguish between a primarily endothelial cell versus an extracellular matrix origin for the increased angiogenesis in TSP2-null mice.
Collapse
Affiliation(s)
- P Bornstein
- Department of Biochemistry, University of Washington, Seattle 98195, USA.
| | | | | | | | | |
Collapse
|
37
|
Belperio JA, Keane MP, Arenberg DA, Addison CL, Ehlert JE, Burdick MD, Strieter RM. CXC chemokines in angiogenesis. J Leukoc Biol 2000. [DOI: 10.1189/jlb.68.1.1] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- John A. Belperio
- Department of Internal Medicine, Division of Pulmonary and Critical Medicine, The University of Michigan Medical School, Ann Arbor
| | - Michael P. Keane
- Department of Internal Medicine, Division of Pulmonary and Critical Medicine, The University of Michigan Medical School, Ann Arbor
| | - Douglas A. Arenberg
- Department of Internal Medicine, Division of Pulmonary and Critical Medicine, The University of Michigan Medical School, Ann Arbor
| | - Christina L. Addison
- Department of Internal Medicine, Division of Pulmonary and Critical Medicine, The University of Michigan Medical School, Ann Arbor
| | - Jan E. Ehlert
- Department of Internal Medicine, Division of Pulmonary and Critical Medicine, The University of Michigan Medical School, Ann Arbor
| | - Marie D. Burdick
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, University of California, Los Angeles School of Medicine, The University of Michigan Medical School, Ann Arbor
| | - Robert M. Strieter
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, University of California, Los Angeles School of Medicine, The University of Michigan Medical School, Ann Arbor
| |
Collapse
|
38
|
Abstract
The growth of human tumors and development of metastases depend on the de novo formation of blood vessels. The formation of new blood vessels is tightly regulated by specific growth factors that target receptor tyrosine kinases (RTKs). Vascular endothelial growth factor (VEGF) and the Flk-1/KDR RTK have been implicated as the key endothelial cell-specific factor signaling pathway required for pathological angiogenesis, including tumor neovascularization. Inhibition of the VEGF tyrosine kinase signaling pathway blocks new blood vessel formation in growing tumors, leading to stasis or regression of tumor growth. Advances in understanding the biology of angiogenesis have led to the development of several therapeutic modalities for the inhibition of the VEGF tyrosine kinase signaling pathway. A number of these modalities are under investigation in clinical studies to evaluate their potential to treat human cancers.
Collapse
Affiliation(s)
- G McMahon
- SUGEN, Inc., South San Francisco, California 94080, USA.
| |
Collapse
|
39
|
McDonnell CO, Hill AD, McNamara DA, Walsh TN, Bouchier-Hayes DJ. Tumour micrometastases: the influence of angiogenesis. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2000; 26:105-15. [PMID: 10744927 DOI: 10.1053/ejso.1999.0753] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Many cancer patients have undetected micrometastatic disease at first presentation which ultimately progresses. Angiogenesis-the development of an independent blood supply-is a key event in the growth of metastases. Improved understanding of the influence of angiogenesis on micrometastatic growth may lead to new therapeutic intervention. METHODS This study examines current concepts of the significance of micrometastases and the role of angiogenesis in their development and destruction. A comprehensive review of the literature on micrometastasis and angiogenesis was performed using the Medline database between 1966 and 1999. CONCLUSIONS Advances in technology have improved our ability to diagnose metastatic disease, but micrometastases in loco-regional lymph nodes and at distant sites can only be detected by sophisticated histological techniques. While the significance of micrometastases remains controversial, there is increasing evidence that micrometastatic status provides useful prognostic information and should be part of standard staging techniques. Anti-angiogenic therapy has the potential to favourably influence management of certain cancers by manipulating a number of key events in the metastatic process.
Collapse
Affiliation(s)
- C O McDonnell
- Royal College of Surgeons in Ireland, Department of Surgery, Dublin 9, Ireland
| | | | | | | | | |
Collapse
|
40
|
Galectin-3 induces endothelial cell morphogenesis and angiogenesis. THE AMERICAN JOURNAL OF PATHOLOGY 2000; 156:899-909. [PMID: 10702407 PMCID: PMC1876842 DOI: 10.1016/s0002-9440(10)64959-0] [Citation(s) in RCA: 332] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Increasing evidence suggests that carbohydrate-binding proteins play an essential role in tumor growth and metastasis. However, conflicting results on their function in the regulation of cell proliferation and differentiation during angiogenesis have been reported. We have examined the role of galectin-3 in the regulation of human umbilical vein endothelial cell proliferation, differentiation, migration, and neovascularization. Galectin-3, a carbohydrate-binding protein, with specificity for type 1 and 11 ABH blood group epitopes and polylactosamine glycan containing cell surface glycoproteins, is the major nonintegrin cellular laminin-binding protein. Because galectin-3 expression was shown to be associated in some tumor systems with metastasis, we questioned whether it induces endothelial cell morphogenesis. Here we show that galectin-3 affects chemotaxis and morphology and stimulates capillary tube formation of HUV-EC-C in vitro and angiogenesis in vivo. Endothelial cell morphogenesis is a carbohydrate-dependent process, as it is neutralized by specific sugars and antibodies. These findings demonstrate that endothelial cell surface carbohydrate recognition event(s) can induce a signaling cascade leading to the differentiation and angiogenesis of endothelial cells.
Collapse
|
41
|
Strieter RM, Addison CL, Ehlert JE, Keane MP, Belperio JA, Burdick MD, Arenberg DA. Use of Immunodeficient Mice for the Evaluation of CXC Chemokines in the Regulation of Tumor-associated Angiogenesis. ILAR J 1999; 40:175-182. [PMID: 11406696 DOI: 10.1093/ilar.40.4.175] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Angiogenesis, defined as the growth of new capillaries from pre-existing vessels, is a pervasive biological phenomenon that is at the core of many physiologic and pathologic processes such as tumor growth. The use of human tumor xenografts in immunodeficient mice has provided significant insight into the biology of angiogenesis as it relates to tumor growth and metastasis. Work reviewed in this article supports the notion that net tumor-derived angiogenesis during tumorigenesis of human tumors is determined, in part, by an imbalance in favor of the overexpression of angiogenic (compared with angiostatic) juxtaposed cysteine residue (CXC) chemokines. This paradigm predicts an environment that favors angiogenesis (tumorigenesis) and supports the potential for spontaneous metastases. The article describes the use of immunodeficient mice as an animal model system for characterizing the qualitative and quantitative presence of these angiogenic and angiostatic CXC chemokines during tumorigenesis, as well as determining their net contribution to human tumorigenesis and metastasis in vivo. Various cancer cell lines have been used and xenografted into immunodeficient mice to create human tumor/mouse chimeras, indicating that an imbalance in the biology of angiogenic versus angiostatic CXC chemokines supports a significant portion of human tumor-derived angiogensis that leads to augmented tumorigenesis and spontaneous metastases. It has also been possible to identify potentially therapeutic novel strategies to manipulate the imbalance of angiogenic (compared with angiostatic) CXC chemokines, which may be directly translational to human disease.
Collapse
Affiliation(s)
- Robert M. Strieter
- Department of Internal Medicine, Division of Pulmonary and Critical Medicine, The University of Michigan Medical School, Ann Arbor, Michigan, USA
| | | | | | | | | | | | | |
Collapse
|
42
|
Ribatti D, Vacca A, Nico B, Quondamatteo F, Ria R, Minischetti M, Marzullo A, Herken R, Roncali L, Dammacco F. Bone marrow angiogenesis and mast cell density increase simultaneously with progression of human multiple myeloma. Br J Cancer 1999; 79:451-5. [PMID: 10027312 PMCID: PMC2362443 DOI: 10.1038/sj.bjc.6690070] [Citation(s) in RCA: 188] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Immunohistochemical, cytochemical and ultrastructural data showing vivid angiogenesis and numerous mast cells (MCs) in the bone marrow of 24 patients with active multiple myeloma (MM) compared with 34 patients with non-active MM and 22 patients with monoclonal gammopathy of undetermined significance (MGUS) led us to hypothesize that angiogenesis parallels progression of MM, and that MCs participate in its induction via angiogenic factors in their secretory granules.
Collapse
Affiliation(s)
- D Ribatti
- Institute of Human Anatomy, Histology and Embryology, University of Bari Medical School, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Vacca A, Ribatti D, Ruco L, Giacchetta F, Nico B, Quondamatteo F, Ria R, Iurlaro M, Dammacco F. Angiogenesis extent and macrophage density increase simultaneously with pathological progression in B-cell non-Hodgkin's lymphomas. Br J Cancer 1999; 79:965-70. [PMID: 10070898 PMCID: PMC2362651 DOI: 10.1038/sj.bjc.6690154] [Citation(s) in RCA: 109] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Node biopsies of 30 benign lymphadenopathies and 71 B-cell non-Hodgkin's lymphomas (B-NHLs) were investigated for microvessel and macrophage counts using immunohistochemistry and morphometric analysis. Both counts were significantly higher in B-NHL. Moreover, when these were grouped into low-grade and high-grade lymphomas, according to the Kiel classification and Working Formulation (WF), statistically significant higher counts were found in the high-grade tumours. Immunohistochemistry and electron microscopy revealed a close spatial association between microvessels and macrophages. Overall, the results suggest that, in analogy to what has already been shown in solid tumours, angiogenesis occurring in B-NHLs increases with tumour progression, and that macrophages promote the induction of angiogenesis via the release of their angiogenic factors.
Collapse
Affiliation(s)
- A Vacca
- Department of Biomedical Sciences and Human Oncology, University of Bari Medical School, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
André T, Chastre E, Kotelevets L, Vaillant JC, Louvet C, Balosso J, Le Gall E, Prévot S, Gespach C. [Tumoral angiogenesis: physiopathology, prognostic value and therapeutic perspectives]. Rev Med Interne 1998; 19:904-13. [PMID: 9887458 DOI: 10.1016/s0248-8663(99)80063-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
INTRODUCTION Angiogenesis activation plays a crucial role in tumoral growth and metastases dissemination. This review summarizes and analyzes current knowledge on molecular mechanisms related to angiogenesis and the prognostic value of its effectors. It also focuses on the therapeutical relevance of various drugs that might inhibit angiogenesic processes. CURRENT KNOWLEDGE AND KEY POINTS Tumor angiogenesis involves complex interactions between tumoral, stromal, endothelial cells, fibroblasts and the extracellular matrix. Normal and malignant angiogenesis depends on the balance of proangiogenic and antiangiogenic factors. Endothelial cells are activated by growth factors, such as Vascular Endothelial Growth Factor (VEGF), and proliferate; they release proteases able to induce degradation of the basement membrane and extracellular matrix, and undergo migration and tubulogenesis. Angiostatin and endostatin are two powerful inhibitors of angiogenesis in experimental models. Assessment of intratumoral microvessel density and quantification of angiogenic factors, including VEGF, are of prognostic value in most cancers, particularly in breast cancer. However, the use of these prognosis markers in clinical practice is still controversial due to the lack of prospective studies and to technical limits inherent to the scoring and standardization of immunohistochemical methods. FUTURE PROSPECTS AND PROJECTS Better understanding of the molecular basis of angiogenesis allows the development of new therapeutical strategies. Biochemical targets of antiangiogenic therapy are: the interaction between angiogenic factors and their receptors; the interaction of endothelial cells with the extracellular matrix; and intracellular signaling pathways. Angiogenesis inhibitors may not cause tumor regression, but inhibit cellular growth and produce "disease dormancy". Extensive phase I to III clinical trials involving antiangiogenesis therapy are in progress.
Collapse
Affiliation(s)
- T André
- Inserm U482, hôpital Saint-Antoine, Paris, France
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Moore BB, Arenberg DA, Addison CL, Keane MP, Strieter RM. Tumor angiogenesis is regulated by CXC chemokines. THE JOURNAL OF LABORATORY AND CLINICAL MEDICINE 1998; 132:97-103. [PMID: 9708570 DOI: 10.1016/s0022-2143(98)90004-x] [Citation(s) in RCA: 43] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- B B Moore
- Department of Internal Medicine, The University of Michigan Medical School, Ann Arbor 48109-0642, USA
| | | | | | | | | |
Collapse
|
46
|
Vacca A, Ribatti D, Iurlaro M, Albini A, Minischetti M, Bussolino F, Pellegrino A, Ria R, Rusnati M, Presta M, Vincenti V, Persico MG, Dammacco F. Human lymphoblastoid cells produce extracellular matrix-degrading enzymes and induce endothelial cell proliferation, migration, morphogenesis, and angiogenesis. INTERNATIONAL JOURNAL OF CLINICAL & LABORATORY RESEARCH 1998; 28:55-68. [PMID: 9594364 DOI: 10.1007/s005990050018] [Citation(s) in RCA: 69] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Human lymphoproliferative diseases can be hypothesized to invade locally and to metastatize via mechanisms similar to those developed by a variety of solid tumors, i.e., the secretion of extracellular matrix-degrading enzymes and stimulation of angiogenesis. To assess this hypothesis, Namalwa, Raji, and Daudi cell lines (Burkitt's lymphoma), LIK and SB cell lines (B-cell lymphoblastic leukemia), CEM and Jurkat cell lines (T-cell lymphoblastic leukemia), and U266 cell line (multiple myeloma) were evaluated for their capacity to produce matrix metalloproteinase-2 and -9, and urokinase-type plasminogen activator. These cell lines were also assessed for their ability: (1) to produce the angiogenic basic fibroblast growth factor and vascular endothelial growth factor; (2) to induce an angiogenic phenotype in cultured endothelial cells, represented by cell proliferation, chemotaxis, and morphogenesis; (3) to stimulate angiogenesis in different in vivo experimental models. All cell lines expressed the mRNA for one or both metalloproteinases. Namalwa, Raji, LIK, SB, and U266 cells secreted the active form of both metalloproteinases, while Daudi, CEM, and Jurkat cells produced metalloproteinase-2 but not-9. In contrast, urokinase-type plasminogen activator was secreted only by SB cells. While Raji, LIK, SB, CEM, and Jurkat cells secreted both basic fibroblast growth factor and vascular endothelial growth factor, Daudi and U266 cells produced only the former, and Namalwa cells only the latter. Accordingly, the conditioned medium of all cell lines stimulated cell proliferation and/or chemotaxis in cultured endothelial cells, with the exception of that of Namalwa cells which was ineffective. The conditioned medium of CEM and Jurkat cells induced morphogenesis in cultured endothelial cells grown on a reconstituted basement membrane (Matrigel). Lastly, Namalwa, Raji, LIK, SB, U266, CEM, and Jurkat cells induced angiogenesis and mononuclear cell recruitment in the murine Matrigel sponge model and in a chick embryo chorioallantoic membrane assay. The extent of angiogenesis in both models was strictly correlated with the density of the mononuclear cell infiltrate. The results indicate that human lymphoproliferative disease cells possess both local and remote invasive ability via the secretion of matrix-degrading enzymes and the induction of angiogenesis which is fostered by host inflammatory cells and by an intervening ensemble of angiogenic factors.
Collapse
Affiliation(s)
- A Vacca
- Department of Biomedical Sciences and Human Oncology, Policlinico, Bari, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Vacca A, Moretti S, Ribatti D, Pellegrino A, Pimpinelli N, Bianchi B, Bonifazi E, Ria R, Serio G, Dammacco F. Progression of mycosis fungoides is associated with changes in angiogenesis and expression of the matrix metalloproteinases 2 and 9. Eur J Cancer 1997; 33:1685-92. [PMID: 9389934 DOI: 10.1016/s0959-8049(97)00186-x] [Citation(s) in RCA: 68] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Changes in angiogenesis and expression of extracellular matrix-degrading enzymes have been substantiated during progression of solid tumours, whereas information on haematological tumours remains circumstantial. In this study, 57 biopsies of mycosis fungoides (MF), a haematological tumour of T-cell lineage, were investigated immunohistochemically for the extent of angiogenesis, and by in situ hybridisation for the expression of matrix metalloproteinases 2 (MMP-2, collagenase A) and 9 (MMP-9, collagenase B). The biopsies we grouped according to the stage of progression: patch-->plaque-->nodular (most advanced). The extent of angiogenesis, as microvessel area, of MF lesions as a whole was significantly higher than that of normal uninjured skin, used as a control. When the stages of MF progression were compared, the values of MF patch stage overlapped that of control skin, while values were significantly higher in the plaque stage and even higher in the nodular stage. In these stages, microvessels were widely scattered in the tumour tissue, in close association with tumour cells, and they frequently displayed arborisation and microaneurysmatic dilation. In contrast, in the patch stage microvessels were irregularly distributed around the tumour aggregates, and arborisation or dilated structures were only rarely seen. The expression of MMP-2 and MMP-9 mRNAs underwent significant upregulation in relation to advancing stage. Indeed, the upstaging was significantly associated with higher proportions of lesions positive for each mRNA or for both, and with lesions with the greatest intensity of expression for each mRNA. Besides tumour cells, the MMP-2 mRNA was expressed by microvascular endothelial cells of intratumour and peri-tumour vessels, and by fibroblasts which were especially abundant in the stroma adjacent to the tumour nodules. The MMP-9 mRNA was found to be present in a subset of tissue macrophages which were more frequently located in close vicinity to the tumour nodules. In contrast, in control skin, a weak positivity for the MMP-2 mRNA in very few microvascular endothelial cells and no signal for the MMP-9 mRNA were observed. These in situ data suggest that angiogenesis and degradation of the extracellular matrix occur simultaneously during MF progression. They imply that interaction between tumour cells and their microvasculature are all the more likely to occur during progression, occasionally with the contribution of tumour-associated stromal cells.
Collapse
Affiliation(s)
- A Vacca
- Department of Biomedical Sciences and Human Oncology, University of Bari Medical School, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|