Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters











Publication year range
1.
Methods Protoc ; 3(4)2020 Nov 21.
Article in English | MEDLINE | ID: mdl-33233345

ABSTRACT

The chick embryo chorioallantoic membrane (CAM) represents a powerful in vivo model to study several physiological and pathological processes including inflammation and tumor progression. Nevertheless, the possibility of deepening the molecular processes in the CAM system is biased by the absence/scarcity of chemical and biological reagents, designed explicitly for avian species. This is particularly true for transcriptional factors, proteinaceous molecules that regulate various cellular responses, including proliferation, survival, and differentiation. Here, we propose a detailed antibody-independent protocol to visualize the activation and nuclear translocation of transcriptional factors in cells or in tissues of different animal species. As a proof of concept, DNA/cAMP response element-binding protein (CREB) interaction was characterized on the CAM tissue using oligonucleotides containing the palindromic binding sequence of CREB. Scrambled oligonucleotides were used as controls. In situ DNA/protein interaction protocol is a versatile method that is useful for the study of transcription factors in the cell and tissue of different origins.

2.
Arterioscler Thromb Vasc Biol ; 34(1): 136-45, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24233491

ABSTRACT

OBJECTIVE: Angiogenesis and inflammation are closely related processes. Gremlin is a novel noncanonical vascular endothelial growth factor receptor-2 (VEGFR2) ligand that induces a proangiogenic response in endothelial cells (ECs). Here, we investigated the role of the cyclic adenosine monophosphate-response element (CRE)-binding protein (CREB) in mediating the proinflammatory and proangiogenic responses of ECs to gremlin. APPROACH AND RESULTS: Gremlin induces a proinflammatory response in ECs, leading to reactive oxygen species and cyclic adenosine monophosphate production and the upregulation of proinflammatory molecules involved in leukocyte extravasation, including chemokine (C-C motif) ligand-2 (Ccl2) and Ccl7, chemokine (C-X-C motif) ligand-1 (Cxcl1), vascular cell adhesion molecule-1 (VCAM-1), and intercellular adhesion molecule-1 (ICAM-1). Accordingly, gremlin induces the VEGFR2-dependent phosphorylation, nuclear translocation, and transactivating activity of CREB in ECs. CREB activation mediates the early phases of the angiogenic response to gremlin, including stimulation of EC motility and permeability, and leads to monocyte/macrophage adhesion to ECs and their extravasation. All these effects are inhibited by EC transfection with a dominant-negative CREB mutant or with a CREB-binding protein-CREB interaction inhibitor that competes for CREB/CRE binding. Also, both recombinant gremlin and gremlin-expressing tumor cells induce proinflammatory/proangiogenic responses in vivo that are suppressed by the anti-inflammatory drug hydrocortisone. Similar effects were induced by the canonical VEGFR2 ligand VEGF-A165. CONCLUSIONS: Together, the results underline the tight cross-talk between angiogenesis and inflammation and demonstrate a crucial role of CREB activation in the modulation of the VEGFR2-mediated proinflammatory/proangiogenic response of ECs to gremlin.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Endothelial Cells/metabolism , Inflammation Mediators/metabolism , Inflammation/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Neovascularization, Physiologic , Active Transport, Cell Nucleus , Angiogenesis Inhibitors/pharmacology , Animals , Anti-Inflammatory Agents/pharmacology , Capillary Permeability , Cell Adhesion , Cell Movement , Chemokine CCL2/metabolism , Chemokine CCL7/metabolism , Chemokine CXCL1/metabolism , Chick Embryo , Coculture Techniques , Culture Media, Conditioned/metabolism , Cyclic AMP/metabolism , Cyclic AMP Response Element-Binding Protein/genetics , Cytokines , Endothelial Cells/drug effects , Endothelial Cells/immunology , Human Umbilical Vein Endothelial Cells/immunology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Hydrocortisone/pharmacology , Inflammation/genetics , Inflammation/immunology , Inflammation/prevention & control , Intercellular Adhesion Molecule-1/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Ligands , MCF-7 Cells , Male , Mice , Mice, Inbred C57BL , Mice, Nude , Neovascularization, Physiologic/drug effects , Phosphorylation , Reactive Oxygen Species/metabolism , Signal Transduction , Time Factors , Transfection , Vascular Cell Adhesion Molecule-1/metabolism , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
3.
Blood ; 116(18): 3677-80, 2010 Nov 04.
Article in English | MEDLINE | ID: mdl-20660291

ABSTRACT

The bone morphogenic protein antagonist gremlin is expressed during embryonic development and under different pathologic conditions, including cancer. Gremlin is a proangiogenic protein belonging to the cystine-knot superfamily that includes transforming growth factor-ß proteins and the angiogenic vascular endothelial growth factors (VEGFs). Here, we demonstrate that gremlin binds VEGF receptor-2 (VEGFR2), the main transducer of VEGF-mediated angiogenic signals, in a bone morphogenic protein-independent manner. Similar to VEGF-A, gremlin activates VEGFR2 in endothelial cells, leading to VEGFR2-dependent angiogenic responses in vitro and in vivo. Gremlin thus represents a novel proangiogenic VEGFR2 agonist distinct from the VEGF family ligands with implications in vascular development, angiogenesis-dependent diseases, and tumor neovascularization.


Subject(s)
Endothelial Cells/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Neoplasms/metabolism , Neovascularization, Pathologic/metabolism , Vascular Endothelial Growth Factor Receptor-2/agonists , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Cattle , Cell Line , Chickens , Endothelial Cells/cytology , Humans , Mice
4.
Blood ; 112(4): 1154-7, 2008 Aug 15.
Article in English | MEDLINE | ID: mdl-18505784

ABSTRACT

Recent observations have shown that Drm, a member the Dan family of bone morphogenic protein (BMP) antagonists, induces endothelial cell (EC) sprouting in vitro and angiogenesis in vivo by interacting with signaling EC receptors in a BMP-independent manner. Here, recombinant Drm (rDrm) up-regulates angiopoientin-1 (Ang-1) expression in EC without affecting Ang-2 and Tie-2 receptor expression. Ang-1 up-regulation is mediated by the activation of the transcription factor NF-kappaB. Specific inhibition of Ang-1 activity by anti-Ang-1 antibodies, soluble Tie-2 receptor, or Ang-1 siRNA transfection significantly reduced the rDrm-mediated sprouting of EC in three-dimensional fibrin and type I collagen gels. In addition, Ang-1 antagonists inhibited the angiogenic activity exerted by rDrm in the chick embryo chorioallantoic membrane. Taken together, the data indicate that the proangiogenic activity of Drm is mediated by the activation of an Ang-1-dependent autocrine loop of stimulation in EC.


Subject(s)
Angiopoietin-1/physiology , Bone Morphogenetic Proteins/antagonists & inhibitors , Intercellular Signaling Peptides and Proteins/physiology , Neovascularization, Physiologic , Angiogenic Proteins , Angiopoietin-1/genetics , Animals , Autocrine Communication , Chick Embryo , Cytokines , Endothelial Cells , Humans , Mice , NF-kappa B/metabolism , Receptor, TIE-2 , Up-Regulation/genetics
5.
J Biol Chem ; 282(27): 19676-84, 2007 Jul 06.
Article in English | MEDLINE | ID: mdl-17456474

ABSTRACT

The extracellular matrix protein osteopontin (OPN) plays a nonredundant role in atherosclerosis and restenosis. Here we investigated the impact of OPN up-regulation in an in vitro model of re-endothelialization after mechanical injury of the endothelial cell monolayer. Murine aortic endothelial (MAE) cells interact via alpha(v) integrins with the integrin-binding Arg-Gly-Asp OPN sequence and adhere to immobilized OPN. On this basis, MAE cells were stably transfected with a wild-type OPN cDNA (OPN-MAE cells), with an OPN mutant lacking the Arg-Gly-Asp sequence (DeltaRGD-OPN-MAE cells), or with vector alone (mock-MAE cells). When compared with mock-MAE and DeltaRGD-OPN-MAE cells, OPN-MAE cells showed a reduced sprouting activity in fibrin gel, a reduced motility in a Boyden chamber assay, and a reduced capacity to repair the wounded monolayer. Accordingly, OPN-MAE cells at the edge of the wound were unable to form membrane ruffles, to reorganize their cytoskeleton, and to activate the focal adhesion kinase and the small GTPase Rac1, key regulators of the cell entry into the first phase of the cell migration cycle. Accordingly, wounded OPN-MAE cells failed to activate the intracellular signals RhoA and ERK1/2, involved in the later phases of the cell migration cycle. Also, parental MAE cells showed reduced re-endothelialization after wounding when seeded on immobilized OPN and exhibited increased adhesiveness to OPN-enriched extracellular matrix. In conclusion, OPN up-regulation impairs re-endothelialization by inhibiting the first phase of the cell migration cycle via alpha(v) integrin engagement by the extracellular matrix-immobilized protein. This may contribute to the adverse effects exerted by OPN in restenosis and atherosclerosis.


Subject(s)
Cell Movement , Endothelial Cells/metabolism , Integrin alphaV/metabolism , MAP Kinase Signaling System , Osteopontin , Animals , Aorta/metabolism , Aorta/pathology , Atherosclerosis/metabolism , Atherosclerosis/pathology , Cell Line, Transformed , Endothelial Cells/pathology , Extracellular Matrix/metabolism , Fibrin , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Graft Occlusion, Vascular/metabolism , Graft Occlusion, Vascular/pathology , MAP Kinase Signaling System/genetics , Mice , Mitogen-Activated Protein Kinase 1 , Mitogen-Activated Protein Kinase 3/metabolism , Mutation , Neuropeptides/metabolism , Osteopontin/genetics , Recombinant Proteins/genetics , Transfection , rac GTP-Binding Proteins/metabolism , rac1 GTP-Binding Protein
6.
Blood ; 109(5): 1834-40, 2007 Mar 01.
Article in English | MEDLINE | ID: mdl-17077323

ABSTRACT

Angiogenesis plays a key role in various physiologic and pathologic conditions, including tumor growth. Drm/gremlin, a member the Dan family of bone morphogenic protein (BMP) antagonists, is commonly thought to affect different processes during growth, differentiation, and development by heterodimerizing various BMPs. Here, we identify Drm/gremlin as a novel proangiogenic factor expressed by endothelium. Indeed, Drm/gremlin was purified to homogeneity from the conditioned medium of transformed endothelial cells using an endothelial-cell sprouting assay to follow protein isolation. Accordingly, recombinant Drm/gremlin stimulates endothelial-cell migration and invasion in fibrin and collagen gels, binds with high affinity to various endothelial cell types, and triggers tyrosine phosphorylation of intracellular signaling proteins. Also, Drm/gremlin induces neovascularization in the chick embryo chorioallantoic membrane. BMP4 does not affect Drm/gremlin interaction with endothelium, and both molecules exert a proangiogenic activity in vitro and in vivo when administered alone or in combination. Finally, Drm/gremlin is produced by the stroma of human tumor xenografts in nude mice, and it is highly expressed in endothelial cells of human lung tumor vasculature when compared with non-neoplastic lung. Our observations point to a novel, previously unrecognized capacity of Drm/gremlin to interact directly with target endothelial cells and to modulate angiogenesis.


Subject(s)
Angiogenesis Inducing Agents/pharmacology , Bone Morphogenetic Proteins/antagonists & inhibitors , Intercellular Signaling Peptides and Proteins/metabolism , Intercellular Signaling Peptides and Proteins/pharmacology , Amino Acid Sequence , Angiogenesis Inducing Agents/chemistry , Angiogenesis Inducing Agents/isolation & purification , Angiogenesis Inducing Agents/metabolism , Animals , Bone Morphogenetic Proteins/metabolism , Cells, Cultured , Chick Embryo , Cytokines , Endothelial Cells/metabolism , Immunohistochemistry , Intercellular Signaling Peptides and Proteins/chemistry , Intercellular Signaling Peptides and Proteins/isolation & purification , Mice , Molecular Sequence Data , Neoplasms/blood supply , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Protein Binding , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
7.
J Immunol ; 175(5): 2788-92, 2005 Sep 01.
Article in English | MEDLINE | ID: mdl-16116163

ABSTRACT

Angiogenesis plays an important role in tissue remodeling and repair during the late phase of inflammation. In the present study, we show that human dendritic cells (DC) that matured in the presence of anti-inflammatory molecules such as calcitriol, PGE2, or IL-10 (alternatively activated DC) selectively secrete the potent angiogenic cytokine vascular endothelial growth factor (VEGF) isoforms VEGF165 and VEGF121. No VEGF production was observed in immature or classically activated DC. Also, the capacity to produce VEGF was restricted to the myeloid DC subset. When implanted in the chick embryo chorioallantoic membrane, alternatively activated DC elicit a marked angiogenic response, which is inhibited by neutralizing anti-VEGF Abs and by the VEGFR-2 inhibitor SU5416. Therefore, alternatively activated DC may contribute to the resolution of the inflammatory reaction by promoting VEGF-induced angiogenesis.


Subject(s)
Dendritic Cells/physiology , Neovascularization, Physiologic , Calcitriol/pharmacology , Humans , Interleukin-10/pharmacology , RNA, Messenger/analysis , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor Receptor-2/physiology
8.
Cytokine Growth Factor Rev ; 16(2): 159-78, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15863032

ABSTRACT

Fibroblast growth factors (FGFs) are a family of heparin-binding growth factors. FGFs exert their pro-angiogenic activity by interacting with various endothelial cell surface receptors, including tyrosine kinase receptors, heparan-sulfate proteoglycans, and integrins. Their activity is modulated by a variety of free and extracellular matrix-associated molecules. Also, the cross-talk among FGFs, vascular endothelial growth factors (VEGFs), and inflammatory cytokines/chemokines may play a role in the modulation of blood vessel growth in different pathological conditions, including cancer. Indeed, several experimental evidences point to a role for FGFs in tumor growth and angiogenesis. This review will focus on the relevance of the FGF/FGF receptor system in adult angiogenesis and its contribution to tumor vascularization.


Subject(s)
Fibroblast Growth Factors/physiology , Neovascularization, Pathologic/physiopathology , Receptors, Fibroblast Growth Factor/physiology , Animals , Endothelial Cells/physiology , Fibroblast Growth Factor 2/physiology , Genetic Therapy , Heparan Sulfate Proteoglycans/physiology , Heparin/physiology , Humans , Ischemia/therapy , Neoplasms/blood supply , Receptors, Vascular Endothelial Growth Factor/physiology , Vascular Endothelial Growth Factor A/physiology
9.
Arterioscler Thromb Vasc Biol ; 25(1): 71-6, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15514208

ABSTRACT

OBJECTIVE: Low-molecular-weight heparin (LMWH) exerts antitumor activity in clinical trials. The K5 polysaccharide from Escherichia coli has the same structure as the heparin precursor. Chemical and enzymatic modifications of K5 polysaccharide lead to the production of biotechnological heparin-like compounds. We investigated the fibroblast growth factor-2 (FGF2) antagonist and antiangiogenic activity of a series of LMW N,O-sulfated K5 derivatives. METHODS AND RESULTS: Surface plasmon resonance analysis showed that LMW-K5 derivatives bind FGF2, thus inhibiting its interaction with heparin immobilized to a BIAcore sensor chip. Interaction of FGF2 with tyrosine-kinase receptors (FGFRs), heparan sulfate proteoglycans (HSPGs), and alpha(v)beta3 integrin is required for biological response in endothelial cells. Similar to LMWH, LMW-K5 derivatives abrogate the formation of HSPG/FGF2/FGFR ternary complexes by preventing FGF2-mediated attachment of FGFR1-overexpressing cells to HSPG-bearing cells and inhibit FGF2-mediated endothelial cell proliferation. However, LMW-K5 derivatives, but not LMWH, also inhibit FGF2/alpha(v)beta3 integrin interaction and consequent FGF2-mediated endothelial cell sprouting in vitro and angiogenesis in vivo in the chick embryo chorioallantoic membrane. CONCLUSIONS: LMW N,O-sulfated K5 derivatives affect both HSPG/FGF2/FGFR and FGF2/alpha(v)beta3 interactions and are endowed with FGF2 antagonist and antiangiogenic activity. These compounds may provide the basis for the design of novel LMW heparin-like angiostatic compounds.


Subject(s)
Angiogenesis Inhibitors/biosynthesis , Escherichia coli/chemistry , Fibroblast Growth Factor 2/antagonists & inhibitors , Genetic Engineering/methods , Heparin, Low-Molecular-Weight/biosynthesis , Polysaccharides, Bacterial/biosynthesis , Angiogenesis Inhibitors/genetics , Animals , Bacterial Capsules , CHO Cells/chemistry , CHO Cells/metabolism , Cattle , Cell Adhesion/physiology , Cell Line , Cell Proliferation/drug effects , Chick Embryo , Chorioallantoic Membrane/drug effects , Cricetinae , Cricetulus , Endothelial Cells/chemistry , Endothelial Cells/metabolism , Escherichia coli/genetics , Fibroblast Growth Factor 2/analogs & derivatives , Fibroblast Growth Factor 2/metabolism , Fibroblast Growth Factors/analogs & derivatives , Fibroblast Growth Factors/metabolism , Heparan Sulfate Proteoglycans/analogs & derivatives , Heparan Sulfate Proteoglycans/deficiency , Heparan Sulfate Proteoglycans/metabolism , Heparin, Low-Molecular-Weight/chemical synthesis , Heparin, Low-Molecular-Weight/genetics , Integrin alphaVbeta3/metabolism , Mice , Neovascularization, Physiologic/drug effects , Polysaccharides, Bacterial/genetics
10.
Blood ; 104(1): 92-9, 2004 Jul 01.
Article in English | MEDLINE | ID: mdl-15031207

ABSTRACT

The long pentraxin PTX3 is a soluble pattern recognition receptor produced by monocytes and endothelial cells that plays a nonredundant role in inflammation. Several pathologic conditions are characterized by local production of both PTX3 and the angiogenic fibroblast growth factor-2 (FGF2). Here, solid-phase binding assays demonstrated that PTX3 binds with high affinity to FGF2 but not to a panel of cytokines and growth factors, including FGF1, FGF4, and FGF8. Accordingly, PTX3 prevented (125)I-FGF2 binding to endothelial cell receptors, leading to specific inhibition of FGF2-induced proliferation. PTX3 hampered also the motogenic activity exerted by endogenous FGF2 on a wounded endothelial cell monolayer. Moreover, PTX3 cDNA transduction in FGF2-transformed endothelial cells inhibited their autocrine FGF2-dependent proliferation and morphogenesis in vitro and their capacity to generate vascular lesions when injected in nude mice. Finally, PTX3 suppressed neovascularization triggered by FGF2 in the chick embryo chorioallantoic membrane with no effect on physiologic angiogenesis. In contrast, the short pentraxin C-reactive protein was a poor FGF2 ligand/antagonist. These results establish the selective binding of a member of the pentraxin superfamily to a growth factor. PTX3/FGF2 interaction may modulate angiogenesis in various physiopathologic conditions driven by inflammation, innate immunity, and/or neoplastic transformation.


Subject(s)
Angiogenesis Inhibitors/physiology , C-Reactive Protein/physiology , Fibroblast Growth Factor 2/metabolism , Neovascularization, Physiologic/drug effects , Serum Amyloid P-Component/physiology , Angiogenesis Inhibitors/genetics , Angiogenesis Inhibitors/pharmacology , Animals , Autocrine Communication/drug effects , Autocrine Communication/physiology , C-Reactive Protein/chemistry , C-Reactive Protein/genetics , C-Reactive Protein/pharmacology , CHO Cells , Cattle , Cell Division/drug effects , Cell Division/physiology , Cell Line , Chick Embryo , Cricetinae , Cricetulus , Cytokines/metabolism , Endothelial Cells/cytology , Endothelial Cells/metabolism , Fibroblast Growth Factor 2/antagonists & inhibitors , Fibroblast Growth Factor 2/genetics , Heparan Sulfate Proteoglycans/metabolism , Humans , Mice , Mice, Nude , Mitogens/metabolism , Neovascularization, Physiologic/physiology , Radioligand Assay , Receptors, Fibroblast Growth Factor/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Serum Amyloid P-Component/chemistry , Serum Amyloid P-Component/genetics , Serum Amyloid P-Component/pharmacology
11.
Blood ; 102(12): 3954-62, 2003 Dec 01.
Article in English | MEDLINE | ID: mdl-12907433

ABSTRACT

The Kit (White) gene encodes the transmembrane receptor of stem cell factor/Kit ligand (KL) and is essential for the normal development/maintenance of pluripotent primordial germ cells (PGCs), hematopoietic stem cells (HSCs), melanoblasts, and some of their descendants. The molecular basis for the transcriptional regulation of Kit during development of these important cell types is unknown. We investigated Kit regulation in hematopoietic cells and PGCs. We identified 6 DNase I hypersensitive sites (HS1-HS6) within the promoter and first intron of the mouse Kit gene and developed mouse lines expressing transgenic green fluorescent protein (GFP) under the control of these regulatory elements. A construct driven by the Kit promoter and including all 6 HS sites is highly expressed during mouse development in Kit+ cells including PGCs and hematopoietic progenitors (erythroid blast-forming units and mixed colony-forming units). In contrast, the Kit promoter alone (comprising HS1) is sufficient to drive low-level GFP expression in PGCs, but unable to function in hematopoietic cells. Hematopoietic expression further requires the addition of the intronproximal HS2 fragment; HS2 also greatly potentiates the activity in PGCs. Thus, HS2 acts as an enhancer integrating transcriptional signals common to 2 developmentally unrelated stem cell/progenitor lineages. Optimal hematopoietic expression further requires HS3-HS6.


Subject(s)
Gene Expression Regulation, Developmental , Genes, Regulator , Germ Cells/cytology , Hematopoietic Stem Cells/cytology , Proto-Oncogene Proteins c-kit/genetics , Animals , Cell Lineage , Cells, Cultured , Deoxyribonuclease I , Embryo, Mammalian , Green Fluorescent Proteins , Hematopoiesis/genetics , Luminescent Proteins/genetics , Mice , Mice, Transgenic , Multipotent Stem Cells/cytology , Promoter Regions, Genetic , Proto-Oncogene Proteins c-kit/biosynthesis , Tissue Distribution
12.
Biochem Biophys Res Commun ; 295(2): 267-75, 2002 Jul 12.
Article in English | MEDLINE | ID: mdl-12150942

ABSTRACT

4-Hydroxynonenal (HNE), a highly reactive product of lipid peroxidation, has an antiproliferative effect in several tumor cell lines and provokes alteration of cell cycle progression in HL-60 cells. HNE down-regulates c-myc expression in K562, HL-60, and MEL cells. This prompted us to study the cascade of phenomena that, starting from the CKIs expression and the phosphorylation of pRb, arrives at the E2F binding to consensus sequence in the P2 promoter of the c-myc gene. Treatment of HL-60 cells with HNE (1 microM) causes a p53-independent increase of p21(WAF1/CIP1) expression, pRb dephosphorylation, a decrease of low molecular weight E2F complexes and an increase of high molecular weight E2F complexes bound to P2 c-myc promoter. E2F4 expression is reduced by HNE treatment as well as the amount of pRb/E2F4 complexes, whereas the amount of pRb/E2F1 complexes is increased. In conclusion, HNE can affect the pRb/E2F pathway by modifying the expression of several genes involved in the control of cell proliferation.


Subject(s)
Aldehydes/pharmacology , Cell Cycle Proteins , DNA-Binding Proteins , Retinoblastoma Protein/metabolism , Transcription Factors/metabolism , Base Sequence , Cyclin-Dependent Kinases/antagonists & inhibitors , DNA Primers , E2F Transcription Factors , E2F1 Transcription Factor , E2F4 Transcription Factor , Enzyme Inhibitors/pharmacology , Genes, myc , HL-60 Cells , Humans , Phosphorylation , Precipitin Tests , Promoter Regions, Genetic
13.
J Hematother Stem Cell Res ; 11(1): 19-32, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11847001

ABSTRACT

Fibroblast growth factors (FGFs) belong to a family of pleiotropic heparin-binding growth factors. They exert multiple functions on various cell types of mesodermal and neuroectodermal origin, affecting cell proliferation, motility, survival, and differentiation. FGF's exert their activity by interacting with tyrosine kinase receptors (FGFRs) and cell-surface heparan sulfate proteoglycans. This article reviews recent studies on the role of the FGF/FGFR system in embryonic hematopoietic development, hematopoiesis, and hematological tumors. FGFs exert both autocrine and paracrine functions in these biological processes by acting on blood cells and their precursors and accessory cells in the bone marrow, including stromal and endothelial cells.


Subject(s)
Fibroblast Growth Factors/physiology , Hematologic Neoplasms/metabolism , Hematopoiesis , Receptors, Fibroblast Growth Factor/physiology , Animals , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/pharmacology , Hematologic Neoplasms/etiology , Hematologic Neoplasms/pathology , Hematopoiesis/drug effects , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/pathology , Humans , Receptors, Fibroblast Growth Factor/genetics , Receptors, Fibroblast Growth Factor/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL