101
|
Benameur T, Tual-Chalot S, Andriantsitohaina R, Martínez MC. PPARalpha is essential for microparticle-induced differentiation of mouse bone marrow-derived endothelial progenitor cells and angiogenesis. PLoS One 2010; 5:e12392. [PMID: 20811625 PMCID: PMC2928272 DOI: 10.1371/journal.pone.0012392] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2009] [Accepted: 07/28/2010] [Indexed: 12/16/2022] Open
Abstract
Background Bone marrow-derived endothelial progenitor cells (EPCs) are critical for neovascularization. We hypothesized that microparticles (MPs), small fragments generated from the plasma membrane, can activate angiogenic programming of EPCs. Methodology/Principal Findings We studied the effects of MPs obtained from wild type (MPsPPARα+/+) and knock-out (MPsPPARα−/−) mice on EPC differentiation and angiogenesis. Bone marrow-derived cells were isolated from WT or KO mice and were cultured in the presence of MPsPPARα+/+ or MPsPPARα−/− obtained from blood of mice. Only MPsPPARα+/+ harboring PPARα significantly increased EPC, but not monocytic, differentiation. Bone marrow-derived cells treated with MPsPPARα+/+ displayed increased expression of pro-angiogenic genes and increased in vivo angiogenesis. MPsPPARα+/+ increased capillary-like tube formation of endothelial cells that was associated with enhanced expressions of endothelial cell-specific markers. Finally, the effects of MPsPPARα+/+ were mediated by NF-κB-dependent mechanisms. Conclusions/Significance Our results underscore the obligatory role of PPARα carried by MPs for EPC differentiation and angiogenesis. PPARα-NF-κB-Akt pathways may play a pivotal stimulatory role for neovascularization, which may, at least in part, be mediated by bone marrow-derived EPCs. Improvement of EPC differentiation may represent a useful strategy during reparative neovascularization.
Collapse
Affiliation(s)
- Tarek Benameur
- CNRS, UMR 6214, Angers, France
- INSERM, U771, Angers, France
- Faculté de Médecine, Université d'Angers, Angers, France
| | - Simon Tual-Chalot
- CNRS, UMR 6214, Angers, France
- INSERM, U771, Angers, France
- Faculté de Médecine, Université d'Angers, Angers, France
| | - Ramaroson Andriantsitohaina
- CNRS, UMR 6214, Angers, France
- INSERM, U771, Angers, France
- Faculté de Médecine, Université d'Angers, Angers, France
| | - María Carmen Martínez
- CNRS, UMR 6214, Angers, France
- INSERM, U771, Angers, France
- Faculté de Médecine, Université d'Angers, Angers, France
- * E-mail:
| |
Collapse
|
102
|
Sottnik JL, Guth AM, Mitchell LA, Dow SW. Minimally invasive assessment of tumor angiogenesis by fine needle aspiration and flow cytometry. Angiogenesis 2010; 13:251-8. [PMID: 20734228 DOI: 10.1007/s10456-010-9182-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2009] [Accepted: 08/03/2010] [Indexed: 01/01/2023]
Abstract
The development of a new, less invasive, and more rapidly implemented method of quantifying endothelial cell density in tumors could facilitate experimental and clinical studies of angiogenesis. Therefore, we evaluated the utility of tumor fine needle aspiration (FNA) coupled with flow cytometry for assessment of tumor angiogenesis. Samples were obtained from cutaneous tumors of mice using FNA, then immunostained and assessed by flow cytometry to determine the number of CD31(+) endothelial cells. Results of the FNA/flow cytometry technique were compared with quantification of tumor microvessel density using immunohistochemistry. The ability of the FNA/cytometry technique to quantify the effects of anti-angiogenic therapy and to monitor changes in tumor angiogenesis over time in individual tumors was also determined. We found that endothelial cell percentages determined in tumor tissue aspirates by flow cytometry correlated well with the percentages of endothelial cells determined in whole tumor digests by flow cytometry and with tumor microvessel density measurements by immunohistochemistry. Moreover, we found that repeated FNA sampling of tumors did not induce endothelial cell changes. Interestingly, by employing repeated FNA sampling of the same tumors we were able to observe a sudden and marked decline in tumor angiogenesis triggered when tumors reached a certain size. Thus, we conclude that the FNA/flow cytometry technique is an efficient, reproducible, and relatively non-invasive method of rapidly assessing tumor angiogenesis, which could be readily applied to evaluation of tumor angiogenesis in clinical settings in humans.
Collapse
Affiliation(s)
- Joseph L Sottnik
- Department of Clinical Sciences, Animal Cancer Center, Colorado State University, Fort Collins, CO 80523-1620, USA
| | | | | | | |
Collapse
|
103
|
Farinha P, Kyle AH, Minchinton AI, Connors JM, Karsan A, Gascoyne RD. Vascularization predicts overall survival and risk of transformation in follicular lymphoma. Haematologica 2010; 95:2157-60. [PMID: 20713461 DOI: 10.3324/haematol.2009.021766] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Follicular lymphoma patients display heterogeneous overall survival and variable risk of transformation. Recent studies have highlighted the role of the microenvironment. The contribution of microvessel density to follicular lymphoma survival remains controversial. We used a quantitative tumor mapping approach to determine whether the degree of vascularization correlated with outcome in a uniformly treated cohort. Whole-tissue sections of diagnostic biopsies from 84 cases were stained for CD34 and tumor-to-vessel-distance that encompassed 90% of the tumor (TVD(90)) was determined using image analysis. Twenty-one cases with lower TVD(90) showed inferior overall survival (P=0.0001) and high risk of transformation (P=0.01). These cases significantly correlated with increased Lymphoma-Associated Macrophages (χ(2)=0.025). In multivariate analysis macrophages content, IPI and TVD(90) were independent predictors of overall survival (P=0.05, P=0.001 and P=0.01, respectively) and IPI and TVD(90) predicted risk of transformation (P=0.008 and P=0.08, respectively). Increased angiogenesis is an independent marker of inferior survival and may promote transformation.
Collapse
Affiliation(s)
- Pedro Farinha
- Pathology, British Columbia Cancer Agency Center for Lymphoid Cancer, Vancouver, Canada
| | | | | | | | | | | |
Collapse
|
104
|
The vascular microenvironment and systemic sclerosis. Int J Rheumatol 2010; 2010. [PMID: 20814552 PMCID: PMC2931393 DOI: 10.1155/2010/362868] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2010] [Revised: 05/28/2010] [Accepted: 07/06/2010] [Indexed: 01/13/2023] Open
Abstract
The role of the vascular microenvironment in the pathogenesis Systemic Sclerosis (SSc) is appreciated clinically as Raynaud's syndrome with capillary nail bed change. This manifestation of vasculopathy is used diagnostically in both limited and diffuse cutaneous subsets of SSc, and is thought to precede fibrosis. The degree of subsequent fibrosis may also be determined by the vascular microenvironment. This paper describes why the vascular microenvironment might determine the degree of end-organ damage that occurs in SSc, with a focus on vascular cell senescence, endothelial progenitor cells (EPC) including multipotential mesenchymal stem cells (MSC), pericytes, and angiogenic monocytes. An explanation of the role of EPC, pericytes, and angiogenic monocytes is important to an understanding of SSc pathogenesis. An evolving understanding of the vascular microenvironment in SSc may allow directed treatment.
Collapse
|
105
|
Sasajima J, Mizukami Y, Sugiyama Y, Nakamura K, Kawamoto T, Koizumi K, Fujii R, Motomura W, Sato K, Suzuki Y, Tanno S, Fujiya M, Sasaki K, Shimizu N, Karasaki H, Kono T, Kawabe JI, Ii M, Yoshiara H, Kamiyama N, Ashida T, Bardeesy N, Chung DC, Kohgo Y. Transplanting normal vascular proangiogenic cells to tumor-bearing mice triggers vascular remodeling and reduces hypoxia in tumors. Cancer Res 2010; 70:6283-92. [PMID: 20631070 DOI: 10.1158/0008-5472.can-10-0412] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Blood vessels deliver oxygen and nutrients to tissues, and vascular networks are spatially organized to meet the metabolic needs for maintaining homeostasis. In contrast, the vasculature of tumors is immature and leaky, resulting in insufficient delivery of nutrients and oxygen. Vasculogenic processes occur normally in adult tissues to repair "injured" blood vessels, leading us to hypothesize that bone marrow mononuclear cells (BMMNC) may be able to restore appropriate vessel function in the tumor vasculature. Culturing BMMNCs in endothelial growth medium resulted in the early outgrowth of spindle-shaped attached cells expressing CD11b/Flt1/Tie2/c-Kit/CXCR4 with proangiogenic activity. Intravenous administration of these cultured vascular proangiogenic cells (VPC) into nude mice bearing pancreatic cancer xenografts and Pdx1-Cre;LSL-Kras(G12D);p53(lox/+) genetically engineered mice that develop pancreatic ductal adenocarcinoma significantly reduced areas of hypoxia without enhancing tumor growth. The resulting vasculature structurally mimicked normal vessels with intensive pericyte coverage. Increases in vascularized areas within VPC-injected xenografts were visualized with an ultrasound diagnostic system during injection of a microbubble-based contrast agent (Sonazoid), indicating a functional "normalization" of the tumor vasculature. In addition, gene expression profiles in the VPC-transplanted xenografts revealed a marked reduction in major factors involved in drug resistance and "stemness" of cancer cells. Together, our findings identify a novel alternate approach to regulate abnormal tumor vessels, offering the potential to improve the delivery and efficacy of anticancer drugs to hypoxic tumors.
Collapse
Affiliation(s)
- Junpei Sasajima
- Division of Gastroenterology and Hematology/Oncology, Department of Medicine, Asahikawa Medical College, Asahikawa, Hokkaido, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
106
|
Zhang W, Zhu XD, Sun HC, Xiong YQ, Zhuang PY, Xu HX, Kong LQ, Wang L, Wu WZ, Tang ZY. Depletion of Tumor-Associated Macrophages Enhances the Effect of Sorafenib in Metastatic Liver Cancer Models by Antimetastatic and Antiangiogenic Effects. Clin Cancer Res 2010; 16:3420-30. [PMID: 20570927 DOI: 10.1158/1078-0432.ccr-09-2904] [Citation(s) in RCA: 310] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
MESH Headings
- Angiogenesis Inhibitors/therapeutic use
- Animals
- Benzenesulfonates/therapeutic use
- Carcinoma, Hepatocellular/blood supply
- Carcinoma, Hepatocellular/drug therapy
- Carcinoma, Hepatocellular/immunology
- Carcinoma, Hepatocellular/pathology
- Cell Line, Tumor
- Clodronic Acid/pharmacology
- Diphosphonates/pharmacology
- Drug Therapy, Combination
- Humans
- Imidazoles/pharmacology
- Liposomes/pharmacology
- Liver Neoplasms, Experimental/blood supply
- Liver Neoplasms, Experimental/drug therapy
- Liver Neoplasms, Experimental/immunology
- Liver Neoplasms, Experimental/pathology
- Lung Neoplasms/prevention & control
- Lung Neoplasms/secondary
- Macrophages/immunology
- Male
- Mice
- Mice, Nude
- Niacinamide/analogs & derivatives
- Phenylurea Compounds
- Pyridines/therapeutic use
- Sorafenib
- Xenograft Model Antitumor Assays
- Zoledronic Acid
Collapse
Affiliation(s)
- Wei Zhang
- Liver Cancer Institute and Zhongshan Hospital, Fudan University, Key Laboratory for Carcinogenesis and Cancer Invasion, The Chinese Ministry of Education, Shanghai, P.R. China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
107
|
Rosuvastatin stimulates clonogenic potential and anti-inflammatory properties of endothelial progenitor cells. Cell Biol Int 2010; 34:709-15. [DOI: 10.1042/cbi20090473] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
108
|
Dalton JE, Maroof A, Owens BMJ, Narang P, Johnson K, Brown N, Rosenquist L, Beattie L, Coles M, Kaye PM. Inhibition of receptor tyrosine kinases restores immunocompetence and improves immune-dependent chemotherapy against experimental leishmaniasis in mice. J Clin Invest 2010; 120:1204-16. [PMID: 20234089 PMCID: PMC2846033 DOI: 10.1172/jci41281] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2009] [Accepted: 01/20/2010] [Indexed: 02/03/2023] Open
Abstract
Receptor tyrosine kinases are involved in multiple cellular processes, and drugs that inhibit their action are used in the clinic to treat several types of cancer. However, the value of receptor tyrosine kinase inhibitors (RTKIs) for treating infectious disease has yet to be explored. Here, we have shown in mice that administration of the broad-spectrum RTKI sunitinib maleate (Sm) blocked the vascular remodeling and progressive splenomegaly associated with experimental visceral leishmaniasis. Furthermore, Sm treatment restored the integrity of the splenic microarchitecture. Although restoration of splenic architecture was accompanied by an increase in the frequency of IFN-gamma+CD4+ T cells, Sm treatment alone was insufficient to cause a reduction in tissue parasite burden. However, preconditioning by short-term Sm treatment proved to be successful as an adjunct therapy, increasing the frequency of IFN-gamma+ and IFN-gamma+TNF+CD4+ T cells, enhancing NO production by splenic macrophages, and providing dose-sparing effects when combined with a first-line immune-dependent anti-leishmanial drug. We propose, therefore, that RTKIs may prove clinically useful as agents to restore immune competence before the administration of chemo- or immunotherapeutic drugs in the treatment of visceral leishmaniasis or other diseases involving lymphoid tissue remodeling, including cancer.
Collapse
Affiliation(s)
- Jane E Dalton
- Centre for Immunology and Infection, Hull York Medical School and Department of Biology, University of York, United Kingdom
| | | | | | | | | | | | | | | | | | | |
Collapse
|
109
|
Current world literature. Curr Opin Endocrinol Diabetes Obes 2010; 17:177-85. [PMID: 20190584 DOI: 10.1097/med.0b013e3283382286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
110
|
Adams RH, Eichmann A. Axon guidance molecules in vascular patterning. Cold Spring Harb Perspect Biol 2010; 2:a001875. [PMID: 20452960 DOI: 10.1101/cshperspect.a001875] [Citation(s) in RCA: 299] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Endothelial cells (ECs) form extensive, highly branched and hierarchically organized tubular networks in vertebrates to ensure the proper distribution of molecular and cellular cargo in the vertebrate body. The growth of this vascular system during development, tissue repair or in disease conditions involves the sprouting, migration and proliferation of endothelial cells in a process termed angiogenesis. Surprisingly, specialized ECs, so-called tip cells, which lead and guide endothelial sprouts, share many feature with another guidance structure, the axonal growth cone. Tip cells are motile, invasive and extend numerous filopodial protrusions sensing growth factors, extracellular matrix and other attractive or repulsive cues in their tissue environment. Axonal growth cones and endothelial tip cells also respond to signals belonging to the same molecular families, such as Slits and Roundabouts, Netrins and UNC5 receptors, Semaphorins, Plexins and Neuropilins, and Eph receptors and ephrin ligands. Here we summarize fundamental principles of angiogenic growth, the selection and function of tip cells and the underlying regulation by guidance cues, the Notch pathway and vascular endothelial growth factor signaling.
Collapse
Affiliation(s)
- Ralf H Adams
- Max-Planck-Institute for Molecular Biomedicine, Department of Tissue Morphogenesis, and University of Münster, Faculty of Medicine, Münster, Germany
| | | |
Collapse
|
111
|
Peddareddigari VG, Wang D, DuBois RN. The tumor microenvironment in colorectal carcinogenesis. CANCER MICROENVIRONMENT : OFFICIAL JOURNAL OF THE INTERNATIONAL CANCER MICROENVIRONMENT SOCIETY 2010; 3:149-66. [PMID: 21209781 PMCID: PMC2990487 DOI: 10.1007/s12307-010-0038-3] [Citation(s) in RCA: 159] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2009] [Accepted: 01/03/2010] [Indexed: 02/07/2023]
Abstract
Colorectal cancer is the second leading cause of cancer-related mortality in the United States. Therapeutic developments in the past decade have extended life expectancy in patients with metastatic disease. However, metastatic colorectal cancers remain incurable. Numerous agents that were demonstrated to have significant antitumor activity in experimental models translated into disappointing results in extending patient survival. This has resulted in more attention being focused on the contribution of tumor microenvironment to the progression of a number of solid tumors including colorectal cancer. A more complete understanding of interactions between tumor epithelial cells and their stromal elements will enhance therapeutic options and improve clinical outcome. Here we will review the role of various stromal components in colorectal carcinogenesis and discuss the potential of targeting these components for the development of future therapeutic agents.
Collapse
Affiliation(s)
- Vijay G. Peddareddigari
- Department of Cancer Biology, The University of Texas, M. D. Anderson Cancer Center, Houston, TX 77030 USA
| | - Dingzhi Wang
- Department of Cancer Biology, The University of Texas, M. D. Anderson Cancer Center, Houston, TX 77030 USA
| | - Raymond N. DuBois
- Department of Cancer Biology, The University of Texas, M. D. Anderson Cancer Center, Houston, TX 77030 USA
- Gastrointestinal Medical Oncology, The University of Texas, M. D. Anderson Cancer Center, Unit 118, 1515 Holcombe Boulevard, Houston, TX 77030-4009 USA
| |
Collapse
|
112
|
Li Calzi S, Neu MB, Shaw LC, Kielczewski JL, Moldovan NI, Grant MB. EPCs and pathological angiogenesis: when good cells go bad. Microvasc Res 2010; 79:207-16. [PMID: 20188747 DOI: 10.1016/j.mvr.2010.02.011] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2009] [Revised: 02/18/2010] [Accepted: 02/18/2010] [Indexed: 01/08/2023]
Abstract
Bone-marrow-derived endothelial progenitor cells (EPCs) contribute to angiogenesis-mediated pathological neovascularization, and recent studies have begun to recognize the biological significance of this contribution. This review will discuss the ability of EPCs to contribute to neovascularization in both physiological and pathological conditions. Circulating EPCs were originally identified in 1997 by Asahara as CD34(+) VEGFR2(+) mononuclear cells. These cells differentiated into an endothelial phenotype, expressed endothelial markers, and incorporated into neovessels at sites of ischemia (Asahara et al., 1997). EPCs provide both instructive (release of pro-angiogenic cytokines) and structural (vessel incorporation and stabilization) functions that contribute to the initiation of neo-angiogenesis. EPC populations can be characterized based on surface markers of freshly isolated cells, or they can be described by their in vitro characteristics once placed in culture. However, a major stumbling block to progress in the field has been the lack of consensus among investigators as to the optimal characterization of EPCs. This review intends to address the role of both EPC classes and evaluate how they interact in the setting of pathological angiogenesis. Since the EPCs may be responsible for turning on the "angiogenic switch," strategies have been employed to keep this switch in the "off" position for diseases like cancer, retinopathy, and wet AMD. The expectation is that EPCs will evolve into clinically useful prognostic and predictive tools in cancer and in ocular diseases associated with pathological neovascularization and that targeting this cell type is a key to successful management of patients suffering from diseases associated with pathological neovascularization.
Collapse
Affiliation(s)
- Sergio Li Calzi
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, Florida, USA
| | | | | | | | | | | |
Collapse
|
113
|
Coffelt SB, Lewis CE, Naldini L, Brown JM, Ferrara N, De Palma M. Elusive identities and overlapping phenotypes of proangiogenic myeloid cells in tumors. THE AMERICAN JOURNAL OF PATHOLOGY 2010; 176:1564-76. [PMID: 20167863 DOI: 10.2353/ajpath.2010.090786] [Citation(s) in RCA: 117] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
It is now established that bone marrow-derived myeloid cells regulate tumor angiogenesis. This was originally inferred from studies of human tumor biopsies in which a positive correlation was seen between the number of tumor-infiltrating myeloid cells, such as macrophages and neutrophils, and tumor microvessel density. However, unequivocal evidence was only provided once mouse models were used to examine the effects on tumor angiogenesis by genetically or pharmacologically targeting myeloid cells. Since then, identifying the exact myeloid cell types involved in this process has proved challenging because of myeloid cell heterogeneity and the expression of overlapping phenotypic markers in tumors. As a result, investigators often simply refer to them now as "bone marrow-derived myeloid cells." Here we review the findings of various attempts to phenotype the myeloid cells involved and discuss the therapeutic implications of correctly identifying-and thus being able to target-this proangiogenic force in tumors.
Collapse
Affiliation(s)
- Seth B Coffelt
- Medical School, University of Sheffield, Sheffield, S10 2RX, UK
| | | | | | | | | | | |
Collapse
|
114
|
Chao H, Hirschi KK. Hemato-vascular origins of endothelial progenitor cells? Microvasc Res 2010; 79:169-73. [PMID: 20149806 DOI: 10.1016/j.mvr.2010.02.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2010] [Accepted: 02/04/2010] [Indexed: 01/07/2023]
Abstract
Numerous studies have suggested the presence of precursor cells in various tissues and organs with potential to differentiate into endothelial and mural cells, and contribute to blood vessel formation in different physiological and pathological circumstances. Although there is still a lack of consensus in the field regarding the origin, and phenotypic and functional characteristics of putative vascular progenitor cell populations, all agree that further studies are needed to fully explore and exploit their great potential as cell therapy for vascular diseases, as modulators of postnatal blood vessel formation, and as disease biomarkers. Herein, we will review the phenotypic and functional characteristics of endothelial progenitor/precursor cell types thought to be derived from the hematopoietic and vascular systems and contribute to postnatal blood vessel formation, and discuss their potential lineage relationships.
Collapse
Affiliation(s)
- Hsu Chao
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA
| | | |
Collapse
|
115
|
Shaked Y, Voest EE. Bone marrow derived cells in tumor angiogenesis and growth: are they the good, the bad or the evil? Biochim Biophys Acta Rev Cancer 2009; 1796:1-4. [PMID: 19703646 DOI: 10.1016/j.bbcan.2009.07.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2008] [Revised: 02/13/2009] [Accepted: 02/20/2009] [Indexed: 01/08/2023]
Abstract
Increasing evidence implicates an important role for a variety of bone marrow derived cells (BMDCs) in tumor angiogenesis and metastatic tumor growth. These cells are derived either from the hematopoietic or mesenchymal cell lineage, and they are distinguished, in part, by the expression of the panhematopoietic marker - CD45. Some of these cell populations can colonize tumors perivascularily, and appear to promote angiogenesis and tumor cell proliferation by paracraine mechanisms, whereas others can contribute "directly" to the growth of tumor vessel capillaries or metastases. In this review we focus in particular on the role of hemangiocytes or recruited bone marrow derived circulating cells (RBCCs) in neovascularization, the contribution of VEGFR1+ hematopoietic stem cells and endothelial precursor cells in metastasis, and the involvement of myeloid derived suppressor CD11b+/Gr-1+ cells in the resistance of tumors to certain antiangiogenic drugs, e.g., VEGF blocking antibodies.
Collapse
Affiliation(s)
- Yuval Shaked
- Molecular Pharmacology, Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel.
| | | |
Collapse
|
116
|
Fang Y, Shen J, Yao M, Beagley KW, Hambly BD, Bao S. Granulocyte-macrophage colony-stimulating factor enhances wound healing in diabetes via upregulation of proinflammatory cytokines. Br J Dermatol 2009; 162:478-86. [PMID: 19799605 DOI: 10.1111/j.1365-2133.2009.09528.x] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
BACKGROUND Chronic ulceration, especially in diabetes, remains a substantial clinical problem. Exogenous granulocyte-macrophage colony-stimulating factor (GM-CSF) is efficacious in the treatment of chronic wound healing in both animal models and patients, but its role in diabetic wounds remains to be explored. Objectives Using a diabetic mouse model, to investigate the role of GM-CSF in wound healing. METHODS Clinical observation, histopathology, immunohistochemistry and cytokine assays. RESULTS There was a significant reduction (50%) in GM-CSF production in the wounds of the diabetics compared with nondiabetics. Exogenous GM-CSF substantially enhanced the wound healing in diabetic mice, accompanied by increased interleukin-6 and monocyte chemoattractant protein-1 production. The elevated cytokines correlated with increased neovascularization, and infiltration of macrophages and neutrophils. GM-CSF showed no beneficial effects in nondiabetic wound healing. CONCLUSIONS Our results provide useful guidelines for the clinical management of chronic ulceration in diabetes.
Collapse
Affiliation(s)
- Y Fang
- Department of Plastic Surgery, School of Medicine, Shanghai Third People's Hospital, Shanghai Jiaotong University, Shanghai, China
| | | | | | | | | | | |
Collapse
|
117
|
Gao D, Mittal V. The role of bone-marrow-derived cells in tumor growth, metastasis initiation and progression. Trends Mol Med 2009; 15:333-43. [PMID: 19665928 DOI: 10.1016/j.molmed.2009.06.006] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2008] [Revised: 06/10/2009] [Accepted: 06/10/2009] [Indexed: 12/14/2022]
Abstract
Emerging evidence from murine models suggests that tumor-specific endocrine factors systemically stimulate the quiescent bone marrow (BM) compartment, resulting in the expansion, mobilization and recruitment of BM progenitor cells. Discrete subsets of tumor-instigated BM-derived progenitor cells support tumor progression and metastasis by regulating angiogenesis, inflammation and immune suppression. Notably, clinical studies have begun to reveal that increased BM recruitment in tumors is associated with poor prognosis. Thus, the BM-derived tumor microenvironment is an attractive therapeutic target, and drugs targeting the components of the microenvironment are currently in clinical trials. Here, we focus on recent advances and emerging concepts regarding the intriguing role of BM-derived cells in tumor growth, metastasis initiation and progression, and we discuss future directions in the context of novel diagnostic and therapeutic opportunities.
Collapse
Affiliation(s)
- Dingcheng Gao
- Department of Cardiothoracic Surgery, Lehman Brothers Lung Cancer Research Center, 1300 York Avenue, 525 East 68th street, New York, New York 10065, USA
| | | |
Collapse
|
118
|
van der Laan AM, Piek JJ, van Royen N. Targeting angiogenesis to restore the microcirculation after reperfused MI. Nat Rev Cardiol 2009; 6:515-23. [DOI: 10.1038/nrcardio.2009.103] [Citation(s) in RCA: 103] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
119
|
Collino F, Revelli A, Massobrio M, Katsaros D, Schmitt-Ney M, Camussi G, Bussolati B. Epithelial-mesenchymal transition of ovarian tumor cells induces an angiogenic monocyte cell population. Exp Cell Res 2009; 315:2982-94. [PMID: 19538958 DOI: 10.1016/j.yexcr.2009.06.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2009] [Revised: 06/05/2009] [Accepted: 06/07/2009] [Indexed: 12/26/2022]
Abstract
Vasculogenesis, or recruitment of progenitors able to differentiate into endothelial-like cells, may provide an important contribution to neovessel formation in tumors. However, the factors involved in the vasculogenic process and in particular the role of the epithelial-mesenchymal transition of tumor cells have not yet been investigated. We found a CD14(+)/KDR(+) angiogenic monocyte population in undifferentiated ovarian tumors, significantly increased in the corresponding tumor metastasis. In vitro, monocyte differentiation into CD14(+)/KDR(+) cells was induced by conditioned media from the primary ovarian tumor cells expressing a mesenchymal phenotype. In contrast, the ovarian tumor cell line SKOV3 expressing an epithelial phenotype was unable to stimulate the differentiation of monocytes into CD14(+)/KDR(+) cells. When an epithelial-mesenchymal transition was induced in SKOV3, they acquired this differentiative ability. Moreover, after mesenchymal transition pleiotrophin expression by SKOV3 was increased and conversely its blockade significantly reduced monocyte differentiation. The obtained CD14(+)/KDR(+) cell population showed the expression of endothelial markers, increased the formation of capillary-like structures by endothelial cells and promoted the migration of ovarian tumor cells in vitro. In conclusion, we showed that the epithelial-mesenchymal transition of ovarian tumor cells induced differentiation of monocytes into the pro-angiogenic CD14(+)/KDR(+) population and thus it may provide a tumor microenvironment that favours vasculogenesis and metastatization of the ovarian cancer.
Collapse
Affiliation(s)
- Federica Collino
- Department of Internal Medicine, Molecular Biotechnology Center, University of Turin, Turin, Italy
| | | | | | | | | | | | | |
Collapse
|
120
|
Horrevoets AJG. Angiogenic monocytes: another colorful blow to endothelial progenitors. THE AMERICAN JOURNAL OF PATHOLOGY 2009; 174:1594-6. [PMID: 19395650 DOI: 10.2353/ajpath.2009.090198] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
This Commentary provides perspective on a related article by Sun-Jin Kim and coworkers (Am J Pathol: 172 AJP08-0819), who assess the contribution of bone marrow-derived cells to tumor angiogenesis in a physiologic, non-myeloablative setting and conclude that the actual angiogenic cell type incorporated in the newly formed vessels is actually monocytes/macrophages.
Collapse
Affiliation(s)
- Anton J G Horrevoets
- VU University Medical Center, Molecular Cell Biology and Immunology, Amsterdam, The Netherlands.
| |
Collapse
|