Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Transl Lung Cancer Res ; 6(3): 295-314, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28713675

ABSTRACT

Malignant pleural mesothelioma is an aggressive tumor of the pleura with an overall poor prognosis. Even with surgical resection, for which only a subset of patients are eligible, long term disease free survival is rare. Standard first-line systemic treatment consists of a platinum analog, an anti-metabolite, and sometimes anti-angiogenic therapy, but there is currently no well-established standard therapy for refractory or relapsed disease. This review focuses on efforts to develop improved systemic therapy for the treatment of malignant pleural mesothelioma (MPM) including cytotoxic systemic therapy, a variety of tyrosine kinase inhibitors and their downstream effector pathways, pharmacologic targeting of the epigenome, novel approaches to target proteins expressed on mesothelioma cells (such as mesothelin), arginine depletion therapy, and the emerging role of immunotherapy. Overall, these studies demonstrate the challenges of improving systemic therapy for MPM and highlight the need to develop therapeutic strategies to control this disease.

2.
Nat Med ; 23(4): 450-460, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28288111

ABSTRACT

Although blood-brain barrier (BBB) compromise is central to the etiology of diverse central nervous system (CNS) disorders, endothelial receptor proteins that control BBB function are poorly defined. The endothelial G-protein-coupled receptor (GPCR) Gpr124 has been reported to be required for normal forebrain angiogenesis and BBB function in mouse embryos, but the role of this receptor in adult animals is unknown. Here Gpr124 conditional knockout (CKO) in the endothelia of adult mice did not affect homeostatic BBB integrity, but resulted in BBB disruption and microvascular hemorrhage in mouse models of both ischemic stroke and glioblastoma, accompanied by reduced cerebrovascular canonical Wnt-ß-catenin signaling. Constitutive activation of Wnt-ß-catenin signaling fully corrected the BBB disruption and hemorrhage defects of Gpr124-CKO mice, with rescue of the endothelial gene tight junction, pericyte coverage and extracellular-matrix deficits. We thus identify Gpr124 as an endothelial GPCR specifically required for endothelial Wnt signaling and BBB integrity under pathological conditions in adult mice. This finding implicates Gpr124 as a potential therapeutic target for human CNS disorders characterized by BBB disruption.


Subject(s)
Blood-Brain Barrier/metabolism , Endothelial Cells/metabolism , Glioblastoma/genetics , Infarction, Middle Cerebral Artery/genetics , Intracranial Hemorrhages/genetics , Receptors, G-Protein-Coupled/genetics , Tight Junctions/metabolism , Animals , Blood-Brain Barrier/ultrastructure , Disease Models, Animal , Endothelial Cells/ultrastructure , Extracellular Matrix/metabolism , Flow Cytometry , Fluorescent Antibody Technique , Glioblastoma/metabolism , Infarction, Middle Cerebral Artery/metabolism , Intracranial Hemorrhages/metabolism , Mice , Mice, Knockout , Microscopy, Electron , Microvessels , Pericytes/ultrastructure , Real-Time Polymerase Chain Reaction , Tight Junctions/ultrastructure , Wnt Signaling Pathway
3.
Curr Probl Cancer ; 40(2-4): 95-105, 2016.
Article in English | MEDLINE | ID: mdl-27839747

ABSTRACT

Despite the remarkable success of endocrine therapy in the treatment of patients with estrogen receptor (ER)- positive breast cancer, not all patients derive benefit from such therapy, or may benefit only temporarily before disease progression or relapse occurs. The value of endocrine therapy, which blocks ER signaling by a variety of strategies, lies in its simplicity, lower toxicity, and better alignment with preserved quality of life, particularly when compared to chemotherapy, which is more toxic and has only modest benefits for many patients with ER-positive breast cancer. It is therefore critical that we discover ways to extend endocrine therapy benefit in patients and prevent therapeutic resistance whenever possible. The tremendous evolution in our understanding of endocrine resistance mechanisms, coupled with the increasing availability of novel agents that target resistance pathways, has led to enhanced treatment approaches for patients with ER-positive breast cancer, primarily through combinations of endocrine agents with a variety of pathway inhibitors. Despite these treatment advances and our changing view of ER-positive breast cancer, there is much work that needs to be done. It remains a problem that we cannot reliably predict which subsets of patients will experience disease relapse or progression on endocrine therapy, and as such, combination strategies with targeted agents have largely been used in unselected patients with ER-positive breast cancer, including those who continue to have endocrine-sensitive disease. Patient selection is a significant issue since most of the targeted therapeutics that we use with endocrine therapy are expensive and can be toxic, and we may be inadvertently overtreating patients whose disease can still be controlled with endocrine therapy alone. In this article, we will review current and future strategies in the treatment of ER-positive breast cancer, as well as the evolving role of targeted therapy in the management of endocrine-resistance.


Subject(s)
Antineoplastic Agents, Hormonal/therapeutic use , Breast Neoplasms/drug therapy , Drug Resistance, Neoplasm , Hormone Antagonists/therapeutic use , Animals , Antineoplastic Agents, Hormonal/pharmacology , Breast Neoplasms/metabolism , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Estradiol/analogs & derivatives , Estradiol/pharmacology , Estradiol/therapeutic use , Female , Fulvestrant , Humans , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptor, ErbB-2/metabolism , Receptors, Estrogen/drug effects , Receptors, Estrogen/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism
5.
Transl Stroke Res ; 3(4): 418-27, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23329986

ABSTRACT

Abnormal endothelial proliferation and angiogenesis may contribute to brain arteriovenous malformation (BAVM) formation. G protein-coupled receptor 124 (GPR124) mediates embryonic CNS angiogenesis; thus we investigated the association of single nucleotide polymorphisms (SNPs) and haplotypes in GPR124 with risk of BAVM. Ten tagging SNPs spanning 39 kb of GPR124 were genotyped in 195 Caucasian BAVM patients and 243 Caucasian controls. SNP and haplotype association with risk of BAVM was screened using χ(2) analysis. Associated variants were further evaluated using multivariable logistic regression, adjusting for age and sex. The minor alleles of 3 GPR124 SNPs adjacent to exon 2 and localized to a 16 kb region of high linkage disequilibrium were associated with reduced risk of BAVM (rs7015566 A, P=0.001; rs7823249 T, P=0.014; rs12676965 C, P=0.007). SNP rs7015566 (intron 1) remained associated after permutation testing (additive model P=0.033). Haplotype analysis revealed a significant overall association (χ(2)=12.55, 4 df, P=0.014); 2 haplotypes (ATCC, P=0.006 and GGCT, P=0.008) were associated with risk of BAVM. We genotyped a known synonymous SNP (rs16887051) in exon 2, however genotype frequency did not differ between cases and controls. Sequencing of conserved GPR124 regions revealed a novel indel polymorphism in intron 2. Immunohistochemistry confirmed GPR124 expression in the endothelium with no qualitative difference in expression between BAVM cases and controls. SNP rs7015566 mapping to intron 1 of GPR124 was associated with BAVM susceptibility among Caucasians. Future work is focused on investigating this gene region.

6.
Science ; 330(6006): 985-9, 2010 Nov 12.
Article in English | MEDLINE | ID: mdl-21071672

ABSTRACT

The orphan G protein-coupled receptor (GPCR) GPR124/tumor endothelial marker 5 is highly expressed in central nervous system (CNS) endothelium. Here, we show that complete null or endothelial-specific GPR124 deletion resulted in embryonic lethality from CNS-specific angiogenesis arrest in forebrain and neural tube. Conversely, GPR124 overexpression throughout all adult vascular beds produced CNS-specific hyperproliferative vascular malformations. In vivo, GPR124 functioned cell-autonomously in endothelium to regulate sprouting, migration, and developmental expression of the blood-brain barrier marker Glut1, whereas in vitro, GPR124 mediated Cdc42-dependent directional migration to forebrain-derived, vascular endothelial growth factor-independent cues. Our results demonstrate CNS-specific angiogenesis regulation by an endothelial receptor and illuminate functions of the poorly understood adhesion GPCR subfamily. Further, the functional tropism of GPR124 marks this receptor as a therapeutic target for CNS-related vascular pathologies.


Subject(s)
Neovascularization, Physiologic , Neural Tube/blood supply , Prosencephalon/blood supply , Receptors, G-Protein-Coupled/metabolism , Animals , Blood Vessels/abnormalities , Blood-Brain Barrier/metabolism , Cell Movement , Embryonic Development , Endothelial Cells/physiology , Endothelium, Vascular/embryology , Endothelium, Vascular/metabolism , Gene Deletion , Glucose Transporter Type 1/metabolism , Mesencephalon/blood supply , Mesencephalon/embryology , Mesencephalon/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Neural Tube/embryology , Neural Tube/metabolism , Prosencephalon/embryology , Prosencephalon/metabolism , Receptors, G-Protein-Coupled/genetics , Rhombencephalon/blood supply , Rhombencephalon/embryology , Rhombencephalon/metabolism , Telencephalon/blood supply , Telencephalon/embryology , Telencephalon/metabolism
7.
Cancer Res ; 69(10): 4527-36, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19401451

ABSTRACT

Vascular endothelial growth factor (VEGF), platelet-derived growth factor (PDGF), and their receptors are important targets in cancer therapy based on angiogenesis inhibition. However, it is unclear whether inhibition of VEGF and PDGF together is more effective than inhibition of either one alone. Here, we used two contrasting tumor models to compare the effects of inhibiting VEGF or PDGF alone, by adenovirally generated soluble receptors, to the effects of inhibiting both together. In RIP-Tag2 tumors, VEGF and PDGF inhibition together reduced tumor vascularity and abundance of pericytes. However, VEGF inhibition reduced tumor vascularity without decreasing pericyte density, and PDGF inhibition reduced pericytes without reducing tumor vascularity. By contrast, in Lewis lung carcinomas (LLC), inhibition of VEGF or PDGF reduced blood vessels and pericytes to the same extent as did inhibition of both together. Similar results were obtained using tyrosine kinase inhibitors AG-013736 and imatinib. In LLC, VEGF expression was largely restricted to pericytes and PDGF was largely restricted to endothelial cells, but, in RIP-Tag2 tumors, expression of both growth factors was more widespread and significantly greater than in LLC. These findings suggest that inhibition of PDGF in LLC reduced pericytes, and then tumor vessels regressed because pericytes were the main source of VEGF. The vasculature of RIP-Tag2 tumors, in which most VEGF is from tumor cells, was more resistant to PDGF inhibition. The findings emphasize the interdependence of pericytes and endothelial cells in tumors and the importance of tumor phenotype in determining the cellular effects of VEGF and PDGF inhibitors on tumor vessels.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Platelet-Derived Growth Factor/physiology , Vascular Endothelial Growth Factor A/physiology , Animals , Antineoplastic Agents/therapeutic use , Axitinib , Benzamides , Humans , Imatinib Mesylate , Imidazoles/therapeutic use , Immunoglobulin Fc Fragments/genetics , Indazoles/therapeutic use , Lung Neoplasms/blood supply , Lung Neoplasms/drug therapy , Mice , Mice, Inbred C57BL , Mice, Transgenic , Piperazines/therapeutic use , Platelet-Derived Growth Factor/antagonists & inhibitors , Pyrimidines/therapeutic use , Reverse Transcriptase Polymerase Chain Reaction , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-1/drug effects , Vascular Endothelial Growth Factor Receptor-1/genetics
8.
Lymphat Res Biol ; 6(3-4): 173-80, 2008.
Article in English | MEDLINE | ID: mdl-19093790

ABSTRACT

The vasculature of the central nervous system (CNS) is highly specialized with a blood-brain-barrier, reciprocal neuroepithelial-endothelial cell interactions and extensive pericyte coverage. Developmentally, numerous important signaling pathways participate in CNS angiogenesis to orchestrate the precise timing and spatial arrangement of the complex CNS vascular network. From a therapeutic standpoint, the CNS vasculature has attracted increased attention since many human ailments, such as stroke, retinopathy, cancer and autoimmune disease are intimately associated with the biology of CNS blood vessels. This review focuses on growth factor pathways that have been shown to be important in developmental CNS vascularization through studies of mouse genetic models and human diseases.


Subject(s)
Central Nervous System/blood supply , Central Nervous System/pathology , Gene Expression Regulation, Developmental , Neovascularization, Physiologic , Animals , Blood Vessels/metabolism , Blood-Brain Barrier , Cell Movement , Humans , Integrins/metabolism , Ligands , Mice , Models, Genetic , Retinal Vessels/metabolism , Signal Transduction , Wnt Proteins/metabolism
9.
Development ; 135(24): 3989-93, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18987025

ABSTRACT

Intronic microRNAs have been proposed to complicate the design and interpretation of mouse knockout studies. The endothelial-expressed Egfl7/miR-126 locus contains miR-126 within Egfl7 intron 7, and angiogenesis deficits have been previously ascribed to Egfl7 gene-trap and lacZ knock-in mice. Surprisingly, selectively floxed Egfl7(Delta) and miR-126(Delta) alleles revealed that Egfl7(Delta/Delta) mice were phenotypically normal, whereas miR-126(Delta/Delta) mice bearing a 289-nt microdeletion recapitulated previously described Egfl7 embryonic and postnatal retinal vascular phenotypes. Regulation of angiogenesis by miR-126 was confirmed by endothelial-specific deletion and in the adult cornea micropocket assay. Furthermore, miR-126 deletion inhibited VEGF-dependent Akt and Erk signaling by derepression of the p85beta subunit of PI3 kinase and of Spred1, respectively. These studies demonstrate the regulation of angiogenesis by an endothelial miRNA, attribute previously described Egfl7 vascular phenotypes to miR-126, and document inadvertent miRNA dysregulation as a complication of mouse knockout strategies.


Subject(s)
MicroRNAs/genetics , Neovascularization, Physiologic/genetics , Proteins/genetics , Animals , Base Sequence , Calcium-Binding Proteins , DNA Primers/genetics , EGF Family of Proteins , Gene Expression Regulation, Developmental , Introns , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , MicroRNAs/metabolism , Phenotype , Proteins/metabolism , Retinal Vessels/embryology , Retinal Vessels/growth & development , Retinal Vessels/metabolism , Sequence Deletion
10.
J Clin Invest ; 116(10): 2610-21, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17016557

ABSTRACT

Inhibitors of VEGF signaling can block angiogenesis and reduce tumor vascularity, but little is known about the reversibility of these changes after treatment ends. In the present study, regrowth of blood vessels in spontaneous RIP-Tag2 tumors and implanted Lewis lung carcinomas in mice was assessed after inhibition of VEGF receptor signaling by AG-013736 or AG-028262 for 7 days. Both agents caused loss of 50%-60% of tumor vasculature. Empty sleeves of basement membrane were left behind. Pericytes also survived but had less alpha-SMA immunoreactivity. One day after drug withdrawal, endothelial sprouts grew into empty sleeves of basement membrane. Vessel patency and connection to the bloodstream followed close behind. By 7 days, tumors were fully revascularized, and the pericyte phenotype returned to baseline. Importantly, the regrown vasculature regressed as much during a second treatment as it did in the first. Inhibition of MMPs or targeting of type IV collagen cryptic sites by antibody HUIV26 did not eliminate the sleeves or slow revascularization. These results suggest that empty sleeves of basement membrane and accompanying pericytes provide a scaffold for rapid revascularization of tumors after removal of anti-VEGF therapy and highlight their importance as potential targets in cancer therapy.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Carcinoma, Lewis Lung/drug therapy , Insulinoma/drug therapy , Neovascularization, Pathologic/drug therapy , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Actins/metabolism , Angiogenesis Inhibitors/pharmacology , Animals , Antibodies, Monoclonal/pharmacology , Axitinib , Basement Membrane/drug effects , Basement Membrane/metabolism , Basement Membrane/pathology , Blood Vessels/drug effects , Blood Vessels/metabolism , Blood Vessels/pathology , Carcinoma, Lewis Lung/blood supply , Carcinoma, Lewis Lung/pathology , Collagen Type IV/immunology , Collagen Type IV/metabolism , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Imidazoles/pharmacology , Imidazoles/therapeutic use , Indazoles/pharmacology , Indazoles/therapeutic use , Insulinoma/blood supply , Insulinoma/pathology , Matrix Metalloproteinase Inhibitors , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasms/blood supply , Neoplasms/drug therapy , Neoplasms/pathology , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Organic Chemicals/pharmacology , Pericytes/drug effects , Pericytes/metabolism , Pericytes/pathology , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Receptor, Platelet-Derived Growth Factor beta/metabolism , Treatment Outcome , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/metabolism
11.
Am J Physiol Heart Circ Physiol ; 290(2): H560-76, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16172168

ABSTRACT

Unlike during development, blood vessels in the adult are generally thought not to require VEGF for normal function. However, VEGF is a survival factor for many tumor vessels, and there are clues that some normal blood vessels may also depend on VEGF. In this study, we sought to identify which, if any, vascular beds in adult mice depend on VEGF for survival. Mice were treated with a small-molecule VEGF receptor (VEGFR) tyrosine kinase inhibitor or soluble VEGFRs for 1-3 wk. Blood vessels were assessed using immunohistochemistry or scanning or transmission electron microscopy. In a study of 17 normal organs after VEGF inhibition, we found significant capillary regression in pancreatic islets, thyroid, adrenal cortex, pituitary, choroid plexus, small-intestinal villi, and epididymal adipose tissue. The amount of regression was dose dependent and varied from organ to organ, with a maximum of 68% in thyroid, but was less in normal organs than in tumors in RIP-Tag2-transgenic mice or in Lewis lung carcinoma. VEGF-dependent capillaries were fenestrated, expressed high levels of both VEGFR-2 and VEGFR-3, and had normal pericyte coverage. Surviving capillaries in affected organs had fewer fenestrations and less VEGFR expression. All mice appeared healthy, but distinct physiological changes, including more efficient blood glucose handling, accompanied some regimens of VEGF inhibition. Strikingly, most capillaries in the thyroid grew back within 2 wk after cessation of treatment for 1 wk. Our findings of VEGF dependency of normal fenestrated capillaries and rapid regrowth after regression demonstrate the plasticity of the adult microvasculature.


Subject(s)
Aging , Capillaries/physiology , Vascular Endothelial Growth Factor A/physiology , Animals , Axitinib , Blood Pressure , Capillaries/ultrastructure , Carcinoma, Lewis Lung/blood supply , Glucose Tolerance Test , Heart/physiology , Imidazoles , Indazoles/pharmacology , Islets of Langerhans/blood supply , Kidney/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasm Transplantation , Pancreatic Neoplasms/blood supply , Phenotype , Reference Values , Regeneration , Signal Transduction/drug effects , Signal Transduction/physiology , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-1/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
12.
Histochem Cell Biol ; 122(5): 477-83, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15558296

ABSTRACT

Endothelial expression of the gap junction proteins, connexin (Cx) 37, Cx40, and Cx43, varies within the vascular network. While previous studies suggest that shear stress may upregulate Cx43, it is not well understood if shear stress affects the expression of all endothelial connexins and to what extent. Endothelial cells on the upstream and downstream surfaces of cardiac valves are subjected to considerably different intensities of shear stress. We therefore reasoned that we could determine the extent hemodynamic forces affect the expression of Cx37, Cx40, and Cx43 by comparing their immunohistochemical distribution on the upstream and downstream surfaces of rat cardiac valves. We found 70- to 200-fold greater expression of Cx43 in the endothelial cells on the upstream than on the downstream surfaces. However, Cx37 was expressed almost equally in the endothelial cells on upstream and downstream surfaces, and Cx40, a major connexin in most vascular endothelial cells, was not detected on either surface. In addition to the heterogeneity in Cx43 expression, endothelial cells on the upstream surface were 35% to 65% smaller than those on the corresponding downstream surface. These results suggest that shear stress may affect endothelial cell size and Cx43 expression but not Cx37 expression.


Subject(s)
Connexin 43/genetics , Endothelium, Vascular/metabolism , Gene Expression Regulation , Heart Valves/cytology , Up-Regulation , Animals , Antibodies, Monoclonal/metabolism , Cell Size , Connexin 43/metabolism , Immunohistochemistry , Rats , Rats, Wistar , Stress, Mechanical
13.
Am J Pathol ; 165(1): 35-52, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15215160

ABSTRACT

Angiogenesis inhibitors are receiving increased attention as cancer therapeutics, but little is known of the cellular effects of these inhibitors on tumor vessels. We sought to determine whether two agents, AG013736 and VEGF-Trap, that inhibit vascular endothelial growth factor (VEGF) signaling, merely stop angiogenesis or cause regression of existing tumor vessels. Here, we report that treatment with these inhibitors caused robust and early changes in endothelial cells, pericytes, and basement membrane of vessels in spontaneous islet-cell tumors of RIP-Tag2 transgenic mice and in subcutaneously implanted Lewis lung carcinomas. Strikingly, within 24 hours, endothelial fenestrations in RIP-Tag2 tumors disappeared, vascular sprouting was suppressed, and patency and blood flow ceased in some vessels. By 7 days, vascular density decreased more than 70%, and VEGFR-2 and VEGFR-3 expression was reduced in surviving endothelial cells. Vessels in Lewis lung tumors, which lacked endothelial fenestrations, showed less regression. In both tumors, pericytes did not degenerate to the same extent as endothelial cells, and those on surviving tumor vessels acquired a more normal phenotype. Vascular basement membrane persisted after endothelial cells degenerated, providing a ghost-like record of pretreatment vessel number and location and a potential scaffold for vessel regrowth. The potent anti-vascular action observed is evidence that VEGF signaling inhibitors do more than stop angiogenesis. Early loss of endothelial fenestrations in RIP-Tag2 tumors is a clue that vessel phenotype may be predictive of exceptional sensitivity to these inhibitors.


Subject(s)
Basement Membrane/drug effects , Endothelium, Vascular/drug effects , Neoplasms/blood supply , Neovascularization, Pathologic , Signal Transduction/drug effects , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Animals , Basement Membrane/pathology , Basement Membrane/ultrastructure , Endothelium, Vascular/pathology , Endothelium, Vascular/ultrastructure , Fluorescein-5-isothiocyanate , Fluorescent Dyes , Immunohistochemistry , Lectins/metabolism , Lung Neoplasms/blood supply , Lung Neoplasms/pathology , Lung Neoplasms/ultrastructure , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Confocal , Microscopy, Electron , Microscopy, Electron, Scanning , Neoplasms/pathology , Neoplasms/ultrastructure , Neovascularization, Pathologic/drug therapy , Vascular Endothelial Growth Factor A/pharmacology
14.
Am J Pathol ; 163(5): 1801-15, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14578181

ABSTRACT

Often described as incomplete or absent, the basement membrane of blood vessels in tumors has attracted renewed attention as a source of angiogenic and anti-angiogenic molecules, site of growth factor binding, participant in angiogenesis, and potential target in cancer therapy. This study evaluated the composition, extent, and structural integrity of the basement membrane on blood vessels in three mouse tumor models: spontaneous RIP-Tag2 pancreatic islet tumors, MCa-IV mammary carcinomas, and Lewis lung carcinomas. Tumor vessels were identified by immunohistochemical staining for the endothelial cell markers CD31, endoglin (CD105), vascular endothelial growth factor receptor-2, and integrin alpha5 (CD49e). Confocal microscopic studies revealed that basement membrane identified by type IV collagen immunoreactivity covered >99.9% of the surface of blood vessels in the three tumors, just as in normal pancreatic islets. Laminin, entactin/nidogen, and fibronectin immunoreactivities were similarly ubiquitous on tumor vessels. Holes in the basement membrane, found by analyzing 1- micro m confocal optical sections, were <2.5 micro m in diameter and involved only 0.03% of the vessel surface. Despite the extensive vessel coverage, the basement membrane had conspicuous structural abnormalities, including a loose association with endothelial cells and pericytes, broad extensions away from the vessel wall, and multiple layers visible by electron microscopy. Type IV collagen-immunoreactive sleeves were also present on endothelial sprouts, supporting the idea that basement membrane is present where sprouts grow and regress. These findings indicate that basement membrane covers most tumor vessels but has profound structural abnormalities, consistent with the dynamic nature of endothelial cells and pericytes in tumors.


Subject(s)
Basement Membrane/pathology , Endothelium, Vascular/pathology , Neovascularization, Pathologic/pathology , Animals , Basement Membrane/ultrastructure , Disease Models, Animal , Endothelium, Vascular/ultrastructure , Immunohistochemistry , Lung Neoplasms/blood supply , Lung Neoplasms/pathology , Male , Mammary Neoplasms, Experimental/blood supply , Mammary Neoplasms, Experimental/pathology , Mice , Microscopy, Confocal , Microscopy, Electron , Pancreatic Neoplasms/blood supply , Pancreatic Neoplasms/pathology
15.
Am J Physiol Heart Circ Physiol ; 285(5): H1917-38, 2003 Nov.
Article in English | MEDLINE | ID: mdl-12842817

ABSTRACT

Profiling gene expression in endothelial cells advances the understanding of normal vascular physiology and disease processes involving angiogenesis. However, endothelial cell purification has been challenging because of the difficulty of isolating cells and their low abundance. Here we examine gene expression in endothelial cells freshly isolated from lung capillaries after in vivo labeling with fluorescent cationic liposomes and purification by fluorescence-activated cell sorting (FACS). Of the 39,000 genes and expressed sequence tags evaluated on custom oligonucleotide arrays, 555 were enriched in endothelial cell fraction. These included familiar endothelial cell-associated genes such as VEGF, VEGF receptor (VEGFR)-1, VEGFR-2, angiopoietin-2, Tie1, Tie2, Edg1 receptor, VE-cadherin, claudin 5, connexin37, CD31, and CD34. Also enriched were genes in semaphorin/neuropilin (Sema3c and Nrp1), ephrin/Eph (ephrin A1, B1, B2, and EphB4), delta/notch (Hey1, Jagged 2, Notch 1, Notch 4, Numb, and Siah1b), and Wingless (Frizzled-4 and Tle1) signaling pathways involved in vascular development and angiogenesis. Expression of representative genes in alveolar capillary endothelial cells was verified by immunohistochemistry. Such expression reflects features that endothelial cells of normal lung capillaries have in common with embryonic and growing blood vessels. About half of the enriched genes, including exostosin 2, lipocalin 7, phospholipid scramblase 2, pleckstrin 2, protocadherin 1, Ryk, scube 1, serpinh1, SNF-related kinase, and several tetraspanins, had little or no previous association with endothelial cells. This approach can readily be used to profile genes expressed in blood vessels in tumors, chronic inflammation, and other sites in which endothelial cells avidly take up cationic liposomes.


Subject(s)
Endothelial Cells/physiology , Neovascularization, Physiologic/genetics , Oligonucleotide Array Sequence Analysis , Pulmonary Circulation/genetics , Age Factors , Animals , Cell Fractionation , Endothelium, Vascular/cytology , Endothelium, Vascular/physiology , Flow Cytometry , Gene Expression/physiology , Immunohistochemistry , Liposomes , Male , Mice , Mice, Inbred C57BL , Specific Pathogen-Free Organisms
SELECTION OF CITATIONS
SEARCH DETAIL
...