Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 255
Filter
Add more filters










Publication year range
1.
Br J Cancer ; 100(6): 865-9, 2009 Mar 24.
Article in English | MEDLINE | ID: mdl-19240721

ABSTRACT

Tumour blood vessels differ from their normal counterparts for reasons that have received little attention. We report here that they are of at least six distinct types, we describe how each forms, and, looking forward, encourage the targeting of tumour vessel subsets that have lost their vascular endothelial growth factor-A (VEGF-A) dependency and so are likely unresponsive to anti-VEGF-A therapies.


Subject(s)
Neoplasms/blood supply , Neovascularization, Pathologic/etiology , Animals , Blood Vessels/pathology , Capillaries/pathology , Capillaries/physiopathology , Humans , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/physiology
2.
J Thromb Haemost ; 3(8): 1835-42, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16102050

ABSTRACT

Angiogenesis has critical roles in normal vascular development and in important pathologies including cancer, wound healing and inflammation. This brief article will review the angiogenic response induced by the vascular permeability factor/vascular endothelial growth factor (VPF/VEGF) family of proteins and particularly VEGF-A, thought to be the single most important angiogenic factor. It will also review the steps and mechanisms by which VEGF-A induces the formation of new blood vessels and will provide an initial classification of the abnormal blood vessels that form in pathological angiogenesis. Finally, it will touch on the exciting relationships that are emerging between angiogenesis and the hemostatic and nervous systems.


Subject(s)
Neovascularization, Pathologic , Neovascularization, Physiologic , Animals , Blood Coagulation , Cell Proliferation , Endothelial Growth Factors/metabolism , Humans , Models, Biological , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism
3.
J Neuropathol Exp Neurol ; 63(8): 841-55, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15330339

ABSTRACT

Angiogenesis is a critical component of stroke, head injury, cerebral vascular malformation development, and brain tumor growth. An understanding of the mechanisms of adult cerebral angiogenesis is fundamental to therapeutic vessel modulation for these diseases. To study angiogenesis in the central nervous system, we injected an adenoviral vector engineered to express vascular endothelial growth factor (VEGF-A164) into adult murine striatum. Vector-infected astrocytes expressed VEGF-A164 resulting in vascular permeability, hemorrhage, and the formation of greatly enlarged "mother" vessels. Subsequently, endothelial cells and pericytes lining mother vessels proliferated and assembled into glomeruloid bodies, complex cellular arrays interspersed by small vessel lumens. As VEGF-A164 expression declined, glomeruloid bodies involuted through apoptotic processes to engender numerous small daughter vessels. Characterized by modestly enlarged lumens with prominent pericyte coverage, daughter vessels were distributed with a density greater than normal cerebral vessels. Daughter vessels remained stable and patent to 16 months and represented the final stage of VEGF-A-induced cerebral angiogenesis. Together, these findings provide a mechanistic understanding of angiogenesis in cerebral disease processes. Furthermore, the long-term stability of daughter vessels in the absence of exogenous VEGF-A164 expression suggests that VEGF-A may enable therapeutic angiogenesis in brain.


Subject(s)
Brain/blood supply , Brain/drug effects , Neovascularization, Physiologic/drug effects , Vascular Endothelial Growth Factor A/administration & dosage , Vascular Endothelial Growth Factor A/genetics , Adenoviridae/genetics , Animals , Astrocytes/metabolism , Astrocytes/virology , Brain/metabolism , Corpus Striatum/blood supply , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Female , Gene Transfer Techniques , Genetic Vectors , Injections, Intraventricular , Mice , Mice, Nude , Neovascularization, Physiologic/physiology , Vascular Endothelial Growth Factor A/biosynthesis
6.
J Biol Chem ; 276(29): 26969-79, 2001 Jul 20.
Article in English | MEDLINE | ID: mdl-11350975

ABSTRACT

Vascular permeability factor (VPF)/vascular endothelial growth factor (VEGF) achieves its multiple functions by activating two receptor tyrosine kinases, Flt-1 (VEGF receptor-1) and KDR (VEGF receptor-2), both of which are selectively expressed on primary vascular endothelium. To dissect the respective signaling pathways and biological functions mediated by these receptors in primary endothelial cells with these two receptors intact, we developed a chimeric receptor system in which the N terminus of the epidermal growth factor receptor was fused to the transmembrane domain and intracellular domain of KDR (EGDR) and Flt-1 (EGLT). We observed that KDR, but not Flt-1, was responsible for VPF/VEGF-induced human umbilical vein endothelial cell (HUVEC) proliferation and migration. Moreover, Flt-1 showed an inhibitory effect on KDR-mediated proliferation, but not migration. We also demonstrated that the inhibitory function of Flt-1 was mediated through the phosphatidylinositol 3-kinase (PI-3K)-dependent pathway because inhibitors of PI-3K as well as a dominant negative mutant of p85 (PI-3K subunit) reversed the inhibition, whereas a constitutively activated mutant of p110 introduced the inhibition to HUVEC-EGDR. We also observed that, in VPF/VEGF-stimulated HUVECs, the Flt-1/EGLT-mediated down-modulation of KDR/EGDR signaling was at or before intracellular Ca(2+) mobilization, but after KDR/EGDR phosphorylation. By mutational analysis, we further identified that the tyrosine 794 residue of Flt-1 was essential for its antiproliferative effect. Taken together, these studies contribute significantly to our understanding of the signaling pathways and biological functions triggered by KDR and Flt-1 and describe a unique mechanism in which PI-3K acts as a mediator of antiproliferation in primary vascular endothelium.


Subject(s)
Cell Division/physiology , Down-Regulation , Endothelium, Vascular/cytology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins/physiology , Receptor Protein-Tyrosine Kinases/physiology , Receptors, Growth Factor/physiology , Signal Transduction/physiology , Base Sequence , Cell Movement/physiology , Cells, Cultured , DNA Primers , Endothelium, Vascular/enzymology , Humans , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Growth Factor/metabolism , Receptors, Vascular Endothelial Growth Factor , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/metabolism , Vascular Endothelial Growth Factor Receptor-1
7.
Nat Med ; 7(5): 569-74, 2001 May.
Article in English | MEDLINE | ID: mdl-11329058

ABSTRACT

Angiogenesis has an essential role in many important pathological and physiological settings. It has been shown that vascular permeability factor/vascular endothelial growth factor (VPF/VEGF), a potent cytokine expressed by most malignant tumors, has critical roles in vasculogenesis and both physiological and pathological angiogenesis. We report here that at non-toxic levels, the neurotransmitter dopamine strongly and selectively inhibited the vascular permeabilizing and angiogenic activities of VPF/VEGF. Dopamine acted through D2 dopamine receptors to induce endocytosis of VEGF receptor 2, which is critical for promoting angiogenesis, thereby preventing VPF/VEGF binding, receptor phosphorylation and subsequent signaling steps. The action of dopamine was specific for VPF/VEGF and did not affect other mediators of microvascular permeability or endothelial-cell proliferation or migration. These results reveal a new link between the nervous system and angiogenesis and indicate that dopamine and other D2 receptors, already in clinical use for other purposes, might have value in anti-angiogenesis therapy.


Subject(s)
Dopamine/pharmacology , Endothelial Growth Factors/physiology , Lymphokines/physiology , Neovascularization, Pathologic , Protein Isoforms/physiology , Animals , Cell Division/drug effects , Cell Movement/drug effects , Cells, Cultured , Endothelial Growth Factors/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Humans , Immunohistochemistry , Lymphokines/metabolism , Mice , Mice, Inbred C3H , Phosphorylation , Protein Binding , Protein Isoforms/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Growth Factor/metabolism , Receptors, Vascular Endothelial Growth Factor , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
8.
Am J Pathol ; 158(3): 1145-60, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11238063

ABSTRACT

Glomeruloid bodies are a defining histological feature of glioblastoma multiforme and some other tumors and vascular malformations. Little is known about their pathogenesis. We injected a nonreplicating adenoviral vector engineered to express vascular permeability factor/vascular endothelial growth factor-164 (VPF/VEGF(164)) into the ears of athymic mice. This vector infected local cells that strongly expressed VPF/VEGF(164) mRNA for 10 to 14 days, after which expression gradually declined. Locally expressed VPF/VEGF(164) induced an early increase in microvascular permeability, leading within 24 hours to edema and deposition of extravascular fibrin; in addition, many pre-existing microvessels enlarged to form thin-walled, pericyte-poor, "mother" vessels. Glomeruloid body precursors were first detected at 3 days as focal accumulations of rapidly proliferating cells in the endothelial lining of mother vessels, immediately adjacent to cells expressing VPF/VEGF(164). Initially, glomeruloid bodies were comprised of endothelial cells but subsequently pericytes and macrophages also participated. As they enlarged by endothelial cell and pericyte proliferation, glomeruloid bodies severely compromised mother vessel lumens and blood flow. Subsequently, as VPF/VEGF(164) expression declined, glomeruloid bodies devolved throughout a period of weeks by apoptosis and reorganization into normal-appearing microvessels. These results provide the first animal model for inducing glomeruloid bodies and indicate that VPF/VEGF(164) is sufficient for their induction and necessary for their maintenance.


Subject(s)
Endothelial Growth Factors/genetics , Lymphokines/genetics , Models, Animal , Neovascularization, Pathologic , Adenoviridae/genetics , Animals , Apoptosis , Capillary Permeability , Cell Division , Cytokines/biosynthesis , Cytokines/genetics , Endothelial Growth Factors/biosynthesis , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Endothelium, Vascular/ultrastructure , Female , In Situ Hybridization , Lymphokines/biosynthesis , Mice , Mice, Nude , Protein Isoforms/biosynthesis , Protein Isoforms/genetics , RNA, Messenger/biosynthesis , Receptor Protein-Tyrosine Kinases/biosynthesis , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Cytokine/biosynthesis , Receptors, Cytokine/genetics , Receptors, Growth Factor/biosynthesis , Receptors, Growth Factor/genetics , Receptors, Vascular Endothelial Growth Factor , Time Factors , Transcription, Genetic , Transgenes , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
9.
Virchows Arch ; 438(1): 13-22, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11213831

ABSTRACT

Angiogenesis is an important factor in the morphological progression and metastasis of many solid tumours. We studied two homogeneous series of myxofibrosarcoma (MFS) and myxoid/round liposarcoma (MRLS), characterised by distinct vascular patterns and correlated the intratumoral microvessel density (IMD) with morphologic progression in both types of sarcoma. In our study, 43 cases of MFS and 42 cases of MRLS were graded according to established diagnostic criteria. For evaluation of IMD, representative sections were stained immunohistochemically for CD31. After selection of "neovascular hot spots", IMD was calculated by measuring the endothelial surface within twenty 200x fields in relation to the total analysed area. In addition to the correlation of IMD with histological grades of malignancy, a correlation of IMD with the inflammatory infiltrate in MFS was done. To determine whether vascular endothelial growth factor (VEGF) and its receptors, KDR and flt-1, may play a role in the progression of both types of sarcomas, we used mRNA in situ hybridisation (ISH) to study VEGF, KDR and flt-1 expression in selected cases. In addition, the expression of thrombospondin-1, which has been reported to inhibit angiogenesis, and of collagen type I was studied using mRNA ISH. Cases of MFS varied histologically from hypocellular, mainly myxoid, neoplasms (low-grade malignant, 18 cases) to intermediate-grade malignant lesions with increased cellularity and mitotic activity (13 cases), and high-grade malignant cases with marked pleomorphism, high proliferative activity and areas of necrosis in many cases (12 cases). Cases of purely low-grade myxoid liposarcoma (16 cases) were characterised by low-cellularity, mucin pooling and plexiform vasculature. In combined MRLS, these hypocellular areas were admixed with hypercellular, round cell areas (5-80% of the analysed tumour area; 23 cases), and in round cell liposarcoma (three cases) rounded tumour cells predominated (>80% of the analysed tumour area). The average IMD in intermediate and high-grade malignant MFS (4.03 and 4.09, respectively) was significantly higher than in low-grade malignant MFS (2.73). Correlation of vascularity with the inflammatory infiltrate in MFS showed increased IMD only in cases with abundant neutrophils; most of these cases were high-grade malignant neoplasms. In contrast, no statistical correlation between morphological progression and IMD was seen in myxoid liposarcoma (6.08), MRLS (6.57) and round cell liposarcoma (4.07). VEGF mRNA was expressed by tumour cells in all histological grades of MFS and MRLS. VEGF receptor mRNA was weakly expressed by endothelia of newly formed blood vessels in both entities. Interestingly, tumour cells of all analysed cases of MFS strongly expressed collagen type I and thrombospondin-1, while these proteins were not detected in tumour cells of MRLS. In conclusion, morphologic tumour progression in MFS is associated with increased IMD, whereas, in MRLS, no such correlation is seen. Whereas VEGF and VEGF receptor mRNA were expressed in both entities, a characteristic expression profile of collagen type I and thrombospondin-1 in MFS emerged. Further studies are necessary to correlate vascularity and clinical course in MFS and MRLS.


Subject(s)
Fibrosarcoma/blood supply , Fibrosarcoma/pathology , Liposarcoma, Myxoid/blood supply , Liposarcoma, Myxoid/pathology , Neovascularization, Pathologic , Adolescent , Adult , Aged , Aged, 80 and over , Collagen/genetics , Disease Progression , Endothelial Growth Factors/genetics , Female , Fibrosarcoma/chemistry , Humans , In Situ Hybridization , Liposarcoma, Myxoid/chemistry , Lymphokines/genetics , Male , Microcirculation/pathology , Middle Aged , Neoplasm Metastasis , Platelet Endothelial Cell Adhesion Molecule-1/analysis , Proto-Oncogene Proteins/genetics , RNA, Messenger/analysis , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Growth Factor/genetics , Receptors, Vascular Endothelial Growth Factor , Thrombospondin 1/genetics , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factor Receptor-1 , Vascular Endothelial Growth Factors
10.
FASEB J ; 15(2): 458-66, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11156961

ABSTRACT

VPF/VEGF acts selectively on the vascular endothelium to enhance permeability, induce cell migration and division, and delay replicative senescence. To understand the changes in gene expression during endothelial senescence, we investigated genes that were differentially expressed in early vs. late passage (senescent) human dermal endothelial cells (HDMEC) using cDNA array hybridization. Early passage HDMEC cultured with or without VPF/VEGF overexpressed 9 and underexpressed 6 genes in comparison with their senescent counterparts. Thymosin beta-10 expression was modulated by VPF/VEGF and was strikingly down-regulated in senescent EC. The beta-thymosins are actin G-sequestering peptides that regulate actin dynamics and are overexpressed in neoplastic transformation. We have also identified senescent EC in the human aorta at sites overlying atherosclerotic plaques. These EC expressed senescence-associated neutral beta-galactosidase and, in contrast to adventitial microvessel endothelium, exhibited weak staining for thymosin beta-10. ISH performed on human malignant tumors revealed strong thymosin beta-10 expression in tumor blood vessels. This is the first report that Tbeta-10 expression is significantly reduced in senescent EC, that VPF/VEGF modulates thymosin beta-10 expression, and that EC can become senescent in vivo. The reduced expression of thymosin beta-10 may contribute to the senescent phenotype by reducing EC plasticity and thus impairing their response to migratory stimuli.


Subject(s)
Arteriosclerosis/pathology , Cellular Senescence/physiology , Endothelial Growth Factors/pharmacology , Endothelium, Vascular/physiology , Lymphokines/pharmacology , Thymosin/genetics , Actins/genetics , Adenocarcinoma/pathology , Aorta, Thoracic/pathology , Arteriosclerosis/physiopathology , Cells, Cultured , Cellular Senescence/drug effects , Colonic Neoplasms/blood supply , DNA, Complementary , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/pathology , Gene Expression Regulation , Humans , Infant, Newborn , Male , Microcirculation/pathology , RNA, Messenger/genetics , Skin/blood supply , Transcription, Genetic , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
11.
Microvasc Res ; 60(2): 112-20, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10964585

ABSTRACT

All-trans-retinoic acid (RA) and other retinoids modulate cell growth and differentiation, generally favoring terminal cell differentiation and inhibiting carcinogenesis. Retinoids are also reported to inhibit angiogenesis and endothelial cell migration, actions that are also anti-carcinogenic. Vascular permeability factor/vascular endothelial growth factor (VPF/VEGF) is a multifunctional cytokine secreted by many tumors. It renders microvessels hyperpermeable to plasma and stimulates endothelial cell migration and division. To investigate further the mechanisms by which RA inhibits angiogenesis, we evaluated the effects of RA on VPF/VEGF-induced angiogenesis and microvascular permeability. RA selectively inhibited the angiogenic response induced by VPF/VEGF, but not that induced by fibroblast growth factor-2 (FGF-2), in the CAM assay. RA and two of its isomers also inhibited the vascular permeabilizing effect of VPF/VEGF but not that induced by histamine. The vascular permeabilization induced by VPF/VEGF and blocked by RA takes place within 1-15 min, too short a time frame for RA to act by modulating transcription through classic retinoid receptors. RA also inhibited VPF/VEGF-induced phosphorylation of PLC-gamma and synthesis of cGMP but actually increased VPF/VEGF binding to cultured endothelial cells. Taken together, these findings indicate that RA selectively blocks VPF/VEGF-induced microvascular permeability and angiogenesis and also identify VPF/VEGF as a major target of RA action. The selectivity of RA's action suggests that other, RA-independent pathways must exist for the angiogenesis induced by FGF-2 and the vascular permeabilizing effect of histamine.


Subject(s)
Antineoplastic Agents/pharmacology , Capillary Permeability/drug effects , Endothelial Growth Factors/pharmacology , Lymphokines/pharmacology , Neovascularization, Physiologic/drug effects , Tretinoin/pharmacology , Animals , Biological Assay , Drug Antagonism , Guinea Pigs , Male , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
12.
Am J Pathol ; 156(6): 2179-83, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10854238

ABSTRACT

The angiopoietins are recently described growth factors for vascular endothelium. The Tie1 and Tie2 receptors are expressed by endothelium. Acquired immune deficiency syndrome (AIDS)-associated Kaposi's sarcoma (KS) and cutaneous angiosarcoma are malignancies of endothelial origin. KS involves primarily the skin and mucosal surfaces and is common in AIDS patients. In an effort to determine whether the angiopoietins and Tie receptors play a role in the pathobiology of angiosarcoma and KS, we studied the expression of angiopoietin-1, angiopoietin-2, angiopoietin-4, Tie1, and Tie2 mRNAs in biopsies of KS from 12 AIDS patients, in biopsies of cutaneous angiosarcoma from two patients, and in control biopsies of normal skin from three volunteers by in situ hybridization. Strong expression of angiopoietin-2, Tie1, and Tie2 mRNAs was detected in the tumor cells of KS and cutaneous angiosarcomas, in contrast to the focal low-level expression in normal skin biopsies. Focal low-level expression of angiopoietin-1 was seen in KS, cutaneous angiosarcomas, and in normal skin. Focal low-level expression of angiopoietin-4 was identified in a minority of KS lesions. These findings suggest that the angiopoietins and Tie receptors may play an important role in the pathobiology of KS and cutaneous angiosarcoma and identify additional potential targets for therapeutic intervention in these vascular malignancies.


Subject(s)
Angiopoietins , Growth Substances/metabolism , Hemangiosarcoma/metabolism , Intercellular Signaling Peptides and Proteins , Neoplasm Proteins/metabolism , Proto-Oncogene Proteins , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Cell Surface/metabolism , Sarcoma, Kaposi/metabolism , Skin Neoplasms/metabolism , Acquired Immunodeficiency Syndrome/complications , Angiopoietin-1 , Angiopoietin-2 , Growth Substances/genetics , Hemangiosarcoma/complications , Humans , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Neoplasm Proteins/genetics , Proteins/genetics , Proteins/metabolism , RNA, Messenger/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Receptor, TIE-1 , Receptor, TIE-2 , Receptors, Cell Surface/genetics , Receptors, TIE , Reference Values , Sarcoma, Kaposi/etiology , Skin Neoplasms/complications
13.
J Histochem Cytochem ; 48(4): 545-56, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10727296

ABSTRACT

Vascular permeability factor/vascular endothelial growth factor (VPF/VEGF) interacts with two high-affinity tyrosine kinase receptors, VEGFR-1 and VEGFR-2, to increase microvascular permeability and induce angiogenesis. Both receptors are selectively expressed by vascular endothelial cells and are strikingly increased in tumor vessels. We used a specific antibody to localize VEGFR-2 (FLK-1, KDR) in microvascular endothelium of normal mouse kidneys and in the microvessels induced by the TA3/St mammary tumor or by infection with an adenoviral vector engineered to express VPF/VEGF. A pre-embedding method was employed at the light and electron microscopic levels using either nanogold or peroxidase as reporters. Equivalent staining was observed on both the luminal and abluminal surfaces of tumor- and adenovirus-induced vascular endothelium, but plasma membranes at interendothelial junctions were spared except at sites connected to vesiculovacuolar organelles (VVOs). VEGFR-2 was also localized to the membranes and stomatal diaphragms of some VVOs. This staining distribution is consistent with a model in which VPF/VEGF increases microvascular permeability by opening VVOs to allow the transendothelial cell passage of plasma and plasma proteins.


Subject(s)
Capillary Permeability , Genetic Vectors/metabolism , Kidney/metabolism , Mammary Neoplasms, Experimental/blood supply , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Growth Factor/metabolism , Adenoviridae/genetics , Animals , Capillaries/metabolism , Endothelium, Vascular/metabolism , Female , Kidney/blood supply , Kidney/ultrastructure , Kidney Glomerulus/blood supply , Kidney Glomerulus/metabolism , Kidney Glomerulus/ultrastructure , Kidney Tubules/blood supply , Kidney Tubules/metabolism , Kidney Tubules/ultrastructure , Mammary Neoplasms, Experimental/metabolism , Mice , Mice, Nude , Microscopy, Electron , Neovascularization, Pathologic , Receptors, Vascular Endothelial Growth Factor
14.
Semin Perinatol ; 24(1): 75-8, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10709865

ABSTRACT

Vasculogenesis, the generation of new blood vessels de novo, and angiogenesis, the formation of new blood vessels from preexisting vessels, are mediated by a number of cytokines and growth factors among which vascular permeability factor/vascular endothelial growth factor (VPF/VEGF) is one of the most important. VPF/VEGF is secreted by many tumor cells, at sites of wound healing and chronic inflammation, and in physiological angiogenesis as in corpus luteum formation. VPF/VEGF is a multifunctional cytokine that interacts with two high affinity tyrosine kinase receptors that are selectively expressed on vascular endothelium. This interaction triggers an angiogenic cascade whose steps, among others, include increased microvascular permeability, leading to deposition of a pro-angiogenic extracellular fibrin matrix, and the formation of mother/daughter vessels.


Subject(s)
Endothelial Growth Factors/physiology , Lymphokines/physiology , Neovascularization, Physiologic/physiology , Animals , Humans , Microcirculation/physiology , Microcirculation/physiopathology , Neovascularization, Pathologic/physiopathology , Protein Isoforms/physiology , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
15.
Lab Invest ; 80(1): 99-115, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10653008

ABSTRACT

Vascular permeability factor/vascular endothelial growth factor (VPF/VEGF) is an angiogenic cytokine with potential for the treatment of tissue ischemia. To investigate the properties of the new blood vessels induced by VPF/VEGF, we injected an adenoviral vector engineered to express murine VPF/VEGF164 into several normal tissues of adult nude mice or rats. A dose-dependent angiogenic response was induced in all tissues studied but was more intense and persisted longer (months) in skin and fat than in heart or skeletal muscle (< or =3 weeks). The initial response (within 18 hours) was identical in all tissues studied and was characterized by microvascular hyperpermeability, edema, deposition of an extravascular fibrin gel, and the formation of enlarged, thin-walled pericyte-poor vessels ("mother" vessels). Mother vessels developed from preexisting microvessels after pericyte detachment and basement membrane degradation. Mother vessels were transient structures that evolved variably in different tissues into smaller daughter vessels, disorganized vessel tangles (glomeruloid bodies), and medium-sized muscular arteries and veins. Vascular structures closely resembling mother vessels and each mother vessel derivative have been observed in benign and malignant tumors, in other examples of pathological and physiological angiogenesis, and in vascular malformations. Together these data suggest that VPF/VEGF has a role in the pathogenesis of these entities. They also indicate that the angiogenic response induced by VPF/VEGF is heterogeneous and tissue specific. Finally, the muscular vessels that developed from mother vessels in skin and perimuscle fat have the structure of collaterals and could be useful clinically in the relief of tissue ischemia.


Subject(s)
Endothelial Growth Factors/physiology , Lymphokines/physiology , Neovascularization, Physiologic/physiology , Animals , Capillary Permeability , Female , Mice , Mice, Inbred BALB C , Mice, Nude , Rats , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
16.
Microvasc Res ; 59(1): 24-37, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10625568

ABSTRACT

Tumor microvessels are hyperpermeable to plasma proteins, a consequence of tumor cell-secreted vascular permeability factor/vascular endothelial growth factor (VPF/VEGF). However, the pathways by which macromolecules extravasate from tumor vessels have been little investigated. To characterize tumor vessels more precisely and to elucidate the pathways by which macromolecules extravasated from them, we studied two well-defined, VPF/VEGF-secreting murine carcinomas, MOT and TA3/St. Whether grown in ascites or solid form, MOT tumors induced large, pericyte-poor "mother" vessels whose lining endothelium developed fenestrae that involved 1.8-5.6% of the surface. Fenestrae developed in parallel with markedly reduced endothelial cell vesiculo-vacuolar organelles (VVOs). TA3/St tumors, which secreted more VPF/VEGF than MOT tumors, elicited mother vessels with unchanged VVOs and without fenestrae. In both tumors, a plasma protein tracer, ferritin, extravasated through VVOs and in MOT tumors ferritin also extravasated through fenestrae. Endothelial gaps were not observed in either tumor. Thus, not all VPF/VEGF-secreting tumors induce fenestrated endothelium. Also, VVOs provide an internal store of membrane that can be transferred to the endothelial cell surface to provide the substantial increase in plasma membrane necessary for mother vessel formation in MOT tumors. Such transfer was apparently unnecessary in TA3/St tumors in which extensive early endothelial cell division provided the increased plasma membrane necessary for forming mother vessels.


Subject(s)
Capillary Permeability/physiology , Carcinoma/blood supply , Endothelium, Vascular/metabolism , Ferritins/metabolism , Mammary Neoplasms, Experimental/blood supply , Microcirculation/pathology , Ovarian Neoplasms/blood supply , Animals , Ascites/metabolism , Carcinoma/metabolism , Carcinoma/pathology , Diaphragm/blood supply , Diaphragm/ultrastructure , Endothelium, Vascular/pathology , Endothelium, Vascular/ultrastructure , Female , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mesentery/blood supply , Mesentery/ultrastructure , Mice , Mice, Inbred Strains , Microcirculation/ultrastructure , Microscopy, Electron , Neoplasm Transplantation , Neovascularization, Pathologic/pathology , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Peritoneum/blood supply , Peritoneum/ultrastructure , Tumor Cells, Cultured
17.
Gene ; 240(1): 157-63, 1999 Nov 15.
Article in English | MEDLINE | ID: mdl-10564822

ABSTRACT

Tumors interact with their environment, reprogramming host cells to induce responses such as angiogenesis, inflammation, immunity and immune suppression. To understand these processes, it is important to identify and isolate new genes whose expression is induced in host tissues in response to tumors. Ascites tumors offer an attractive model for isolating such genes, because responding host peritoneal lining tissues can be cleanly separated from tumor cells growing in suspension within the peritoneal cavity. We here report the cloning by differential display of a novel gene, DLM-1, that is highly up-regulated in the peritoneal lining tissue of mice bearing MOT ascites tumors. Mouse peritoneal macrophages, stimulated by IFN-gamma or LPS, also expressed significant amounts of DLM-1. Up-regulation of DLM-1 became evident by 4h after stimulation with IFN-gamma and was not blocked by cycloheximide, suggesting the presence of IFN responding elements in its transcription regulation region. DLM-1 RNA was detected at significant levels in normal mouse lung, intestinal epithelium, liver and thymus by Northern blot analysis. In situ hybridization of MOT and HT-29 mouse subcutaneous transplanted solid tumors revealed strong DLM-1 expression in the host reactive stromal cells, but not the tumor cells. Sequence analysis of the full-length cDNA clone revealed that it encodes a protein of approx. M(r) 44330 with multiple potential protein kinase C and casein kinase II phosphorylation sites. Our data suggest that DLM-1 plays a role in such important processes as host response in neoplasia.


Subject(s)
DNA-Binding Proteins , Glycoproteins/genetics , Macrophage Activation/genetics , Macrophages, Peritoneal/metabolism , RNA, Neoplasm/genetics , Amino Acid Sequence , Animals , Base Sequence , Blotting, Northern , Cloning, Molecular , Cytokines/pharmacology , DNA, Complementary/chemistry , DNA, Complementary/genetics , DNA, Complementary/isolation & purification , Female , Gene Expression Regulation/drug effects , Humans , In Situ Hybridization , Macrophages, Peritoneal/cytology , Macrophages, Peritoneal/drug effects , Male , Mice , Mice, Inbred Strains , Molecular Sequence Data , Neoplasm Transplantation , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins , Sequence Analysis, DNA , Tissue Distribution , Tumor Cells, Cultured , Up-Regulation
18.
Clin Cancer Res ; 5(5): 1041-56, 1999 May.
Article in English | MEDLINE | ID: mdl-10353737

ABSTRACT

The generation of vascular stroma is essential for solid tumor growth and involves stimulatory and inhibiting factors as well as stromal components that regulate functions such as cellular adhesion, migration, and gene expression. In an effort to obtain a more integrated understanding of vascular stroma formation in breast carcinoma, we examined expression of the angiogenic factor vascular permeability factor (VPF)/vascular endothelial growth factor (VEGF); the VPF/VEGF receptors flt-1 and KDR; thrombospondin-1, which has been reported to inhibit angiogenesis; and the stromal components collagen type I, total fibronectin, ED-A+ fibronectin, versican, and decorin by mRNA in situ hybridization on frozen sections of 113 blocks of breast tissue from 68 patients including 28 sections of breast tissue without malignancy, 18 with in situ carcinomas, 56 with invasive carcinomas, and 8 with metastatic carcinomas. A characteristic expression profile emerged that was remarkably similar in invasive carcinoma, carcinoma in situ, and metastatic carcinoma, with the following characteristics: strong tumor cell expression of VPF/VEGF; strong endothelial cell expression of VPF/VEGF receptors; strong expression of thrombospondin-1 by stromal cells and occasionally by tumor cells; and strong stromal cell expression of collagen type I, total fibronectin, ED-A+ fibronectin, versican, and decorin. The formation of vascular stroma preceded invasion, raising the possibility that tumor cells invade not into normal breast stroma but rather into a richly vascular stroma that they have induced. Similarly, tumor cells at sites of metastasis appear to induce the vascular stroma in which they grow. We conclude that a distinct pattern of mRNA expression characterizes the generation of vascular stroma in breast cancer and that the formation of vascular stroma may play a role not only in growth of the primary tumor but also in invasion and metastasis.


Subject(s)
Breast Neoplasms/blood supply , Carcinoma in Situ/blood supply , Carcinoma/blood supply , Neovascularization, Pathologic , Adenocarcinoma, Mucinous/blood supply , Adenocarcinoma, Mucinous/chemistry , Adenocarcinoma, Mucinous/pathology , Biomarkers , Biopsy , Breast/blood supply , Breast/chemistry , Breast Neoplasms/chemistry , Breast Neoplasms/pathology , Carcinoma/chemistry , Carcinoma/pathology , Carcinoma in Situ/chemistry , Carcinoma, Ductal, Breast/blood supply , Carcinoma, Ductal, Breast/chemistry , Carcinoma, Ductal, Breast/pathology , Carcinoma, Intraductal, Noninfiltrating/blood supply , Carcinoma, Intraductal, Noninfiltrating/chemistry , Carcinoma, Intraductal, Noninfiltrating/pathology , Carcinoma, Lobular/blood supply , Carcinoma, Lobular/chemistry , Carcinoma, Lobular/pathology , Chondroitin Sulfate Proteoglycans/analysis , Collagen/analysis , Decorin , Endothelial Growth Factors/analysis , Endothelium, Vascular/chemistry , Epithelial Cells/chemistry , Extracellular Matrix Proteins , Female , Fibrocystic Breast Disease/metabolism , Fibrocystic Breast Disease/pathology , Fibronectins/analysis , Frozen Sections , Humans , In Situ Hybridization , Lectins, C-Type , Lymphatic Metastasis , Lymphokines/analysis , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Proteins/analysis , Protein Isoforms/analysis , Proteoglycans/analysis , Proto-Oncogene Proteins/analysis , RNA, Messenger/analysis , RNA, Neoplasm/analysis , Receptor Protein-Tyrosine Kinases/analysis , Receptors, Growth Factor/analysis , Receptors, Vascular Endothelial Growth Factor , Stromal Cells/pathology
19.
Microcirculation ; 6(1): 23-44, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10100187

ABSTRACT

OBJECTIVE: The goal of these studies was to define the anatomic pathways by which circulating macromolecules extravasate from the hyperpermeable microvessels that supply tumors and from normal venules that have been rendered hyperpermeable by vasoactive mediators. METHODS: Extravasation pathways of circulating macromolecular tracers were followed by several morphological techniques: light and fluorescence microscopy, transmission electron microscopy of routine as well as ultrathin and serial sections, computer-assisted three-dimensional reconstructions, and morphometry. RESULTS AND DISCUSSION: Macromolecules extravasated across tumor microvessels or across normal venules rendered hyperpermeable by VPF/VEGF, histamine, or serotonin by three primary pathways: 1) Vesiculo-vacuolar organelles (VVOs), clusters of cytoplasmic vesicles and vacuoles that span endothelial cytoplasm from lumen to ablumen; 2) trans-endothelial cell (EC), pores, and 3) fenestrae. We also present data concerning the structure and function of VVOs as well as evidence that VVOs form as the result of linking together and fusion of caveolae-sized unit vesicles. Under suitable conditions VVOs also afforded a pathway for macromolecular transport in the reverse direction, i.e., from vascular ablumen to lumen. Finally, in addition to opening VVOs to the passage of macromolecules, mediators such as VPF/VEGF may also induce structural rearrangements of VVOs, transforming them into trans-EC pores or fenestrae.


Subject(s)
Capillary Permeability/drug effects , Endothelial Growth Factors/pharmacology , Endothelium, Vascular/drug effects , Lymphokines/pharmacology , Animals , Endothelium, Vascular/ultrastructure , Humans , Image Processing, Computer-Assisted , Microcirculation , Microscopy, Electron , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
20.
J Histochem Cytochem ; 47(2): 159-67, 1999 Feb.
Article in English | MEDLINE | ID: mdl-9889252

ABSTRACT

In situ vascular endothelium is characterized by many cytoplasmic vesicles (caveolae) and vacuoles. In venules these are organized into prominent clusters called vesiculo-vacuolar organelles or VVOs. VVOs provide an important pathway for plasma protein extravasation in response to vasoactive mediators. In contrast, cultured endothelial cells isolated from many sources lack VVOs and generally have few caveolae. Our goal was to preserve VVOs in cultured endothelium. Bovine adrenal microvascular endothelial cells (BCEs) cultured on floating Matrigel-collagen Type I gels with vascular permeability factor/vascular endothelial growth factor (VPF/VEGF) exhibited typical VVOs by electron microscopy. Both in vivo and in culture VVOs were caveolin-positive by immunoelectron microscopy. On the basis of caveolin immunostaining, VVOs could also be detected by light (confocal) microscopy. When BCEs were cultured without VPF/VEGF, caveolin staining was finely punctate and electron microscopy confirmed the near absence of VVOs. BCE VVOs were sensitive to N-ethylmaleimide. Other types of endothelium cultured on Matrigel-collagen gels with or without VPF/VEGF exhibited few caveolae and no VVOs. Therefore, preservation of VVOs in cultured endothelium required a specific combination of endothelial cells (BCEs), surface matrix (Matrigel-collagen), and growth factor (VPF/VEGF). These endothelial cells should be useful for in vitro studies of trans-endothelial transport.


Subject(s)
Caveolins , Endothelial Growth Factors/pharmacology , Endothelium, Vascular/ultrastructure , Lymphokines/pharmacology , Vacuoles/drug effects , Animals , Cattle , Caveolin 1 , Cell Culture Techniques , Cell Line , Collagen , Drug Combinations , Endothelium, Vascular/chemistry , Endothelium, Vascular/drug effects , Fluorescent Antibody Technique , Laminin , Membrane Proteins/analysis , Microscopy, Electron , Models, Biological , Proteoglycans , Vacuoles/chemistry , Vacuoles/ultrastructure , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors , Veins/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...