Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Dis Model Mech ; 16(10)2023 10 01.
Article in English | MEDLINE | ID: mdl-37718909

ABSTRACT

Sezary syndrome (SS) is a rare, aggressive leukemic variant of cutaneous T-cell lymphoma (CTCL) that lacks adequate therapeutic options and representative small-animal models. Here, we demonstrate that IL-15 is a critical CTCL growth factor. Importantly, an immunodeficient knock-in mouse model genetically engineered to express human IL-15 uniquely supported the growth of SS patient samples relative to conventional immunodeficient mouse strains. SS patient-derived xenograft (PDX) models recapacitated key pathological features of the human disease, including skin infiltration and spread of leukemic cells to the periphery, and maintained the dependence on human IL-15 upon serial in vivo passaging. Detailed molecular characterization of the engrafted cells by single-cell transcriptomic analysis revealed congruent neoplastic gene expression signatures but distinct clonal engraftment patterns. Overall, we document an important dependence of Sezary cell survival and proliferation on IL-15 signaling and the utility of immunodeficient humanized IL-15 mice as hosts for SS - and potentially other T and NK cell-derived hematologic malignancies - PDX model generation. Furthermore, these studies advocate the thorough molecular understanding of the resultant PDX models to maximize their translational impact.


Subject(s)
Lymphoma, T-Cell, Cutaneous , Sezary Syndrome , Skin Neoplasms , Humans , Animals , Mice , Skin Neoplasms/metabolism , Interleukin-15 , Lymphoma, T-Cell, Cutaneous/pathology , Sezary Syndrome/metabolism , Sezary Syndrome/pathology , Lymphocytes/metabolism , Tumor Microenvironment
2.
Sci Transl Med ; 15(702): eadd1175, 2023 06 28.
Article in English | MEDLINE | ID: mdl-37379368

ABSTRACT

Notch signaling promotes T cell pathogenicity and graft-versus-host disease (GVHD) after allogeneic hematopoietic cell transplantation (allo-HCT) in mice, with a dominant role for the Delta-like Notch ligand DLL4. To assess whether Notch's effects are evolutionarily conserved and to identify the mechanisms of Notch signaling inhibition, we studied antibody-mediated DLL4 blockade in a nonhuman primate (NHP) model similar to human allo-HCT. Short-term DLL4 blockade improved posttransplant survival with durable protection from gastrointestinal GVHD in particular. Unlike prior immunosuppressive strategies tested in the NHP GVHD model, anti-DLL4 interfered with a T cell transcriptional program associated with intestinal infiltration. In cross-species investigations, Notch inhibition decreased surface abundance of the gut-homing integrin α4ß7 in conventional T cells while preserving α4ß7 in regulatory T cells, with findings suggesting increased ß1 competition for α4 binding in conventional T cells. Secondary lymphoid organ fibroblastic reticular cells emerged as the critical cellular source of Delta-like Notch ligands for Notch-mediated up-regulation of α4ß7 integrin in T cells after allo-HCT. Together, DLL4-Notch blockade decreased effector T cell infiltration into the gut, with increased regulatory to conventional T cell ratios early after allo-HCT. Our results identify a conserved, biologically unique, and targetable role of DLL4-Notch signaling in intestinal GVHD.


Subject(s)
Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Mice , Humans , Animals , Transplantation, Homologous , Receptors, Notch/metabolism , Signal Transduction , Graft vs Host Disease/metabolism , Primates
3.
Dis Model Mech ; 12(9)2019 09 16.
Article in English | MEDLINE | ID: mdl-31399482

ABSTRACT

T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive hematologic cancer derived from the malignant transformation of T-cell progenitors. Outcomes remain poor for T-ALL patients who have either primary resistance to standard-of-care chemotherapy or disease relapse. Notably, there are currently no targeted therapies available in T-ALL. This lack of next-generation therapies highlights the need for relevant preclinical disease modeling to identify and validate new targets and treatment approaches. Here, we adapted a spontaneously arising, genetically heterogeneous, thymic transplantation-based murine model of T-ALL, recapitulating key histopathological and genetic features of the human disease, to the preclinical testing of targeted and immune-directed therapies. Genetic engineering of the murine Notch1 locus aligned the spectrum of Notch1 mutations in the mouse model to that of human T-ALL and confirmed aberrant, recombination-activating gene (RAG)-mediated 5' Notch1 recombination events as the preferred pathway in murine T-ALL development. Testing of Notch1-targeting therapeutic antibodies demonstrated T-ALL sensitivity to different classes of Notch1 blockers based on Notch1 mutational status. In contrast, genetic ablation of Notch3 did not impact T-ALL development. The T-ALL model was further applied to the testing of immunotherapeutic agents in fully immunocompetent, syngeneic mice. In line with recent clinical experience in T-cell malignancies, programmed cell death 1 (PD-1) blockade alone lacked anti-tumor activity against murine T-ALL tumors. Overall, the unique features of the spontaneous T-ALL model coupled with genetic manipulations and the application to therapeutic testing in immunocompetent backgrounds will be of great utility for the preclinical evaluation of novel therapies against T-ALL.


Subject(s)
Immunotherapy , Molecular Targeted Therapy , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/therapy , Receptors, Notch/metabolism , Signal Transduction , Animals , Antibodies, Monoclonal/metabolism , B7-H1 Antigen/metabolism , Disease Models, Animal , Gene Deletion , Humans , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics , Oncogenes , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Thymus Gland/transplantation
4.
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
5.
Cancer Res ; 75(19): 4086-96, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26377940

ABSTRACT

The Notch ligand delta-like 4 (Dll4) has been identified as a promising target in tumor angiogenesis in preclinical studies, and Dll4 inhibitors have recently entered clinical trials for solid tumors, including ovarian cancers. In this study, we report the development of REGN421 (enoticumab), a fully human IgG1 monoclonal antibody that binds human Dll4 with sub-nanomolar affinity and inhibits Notch signaling. Administering REGN421 to immunodeficient mice engineered to express human Dll4 inhibited the growth of several human tumor xenografts in association with the formation of nonfunctional tumor blood vessels. In ovarian tumor xenograft models, Dll4 was expressed specifically by the tumor endothelium, and Dll4 blockade by human-specific or mouse-specific Dll4 antibodies exerted potent antitumor activity, which relied entirely on targeting Dll4 expressed by tumor stromal cells but not by the tumor cells themselves. However, Dll4 blockade reduced Notch signaling in both blood vessels and tumor cells surrounding the blood vessels, suggesting that endothelial-expressed Dll4 might induce Notch signaling in adjacent ovarian tumor cells. The antitumor effects of targeting Dll4 were augmented significantly by simultaneous inhibition of VEGF signaling, whereas this combined blockade reversed normal organ vascular changes induced by Dll4 blockade alone. Overall, our findings deepen the rationale for antibody-based strategies to target Dll4 in ovarian cancers, especially in combination with VEGF blockade.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Intercellular Signaling Peptides and Proteins/physiology , Molecular Targeted Therapy , Neoplasm Proteins/antagonists & inhibitors , Neovascularization, Pathologic/drug therapy , Ovarian Neoplasms/drug therapy , Adaptor Proteins, Signal Transducing , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/therapeutic use , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Antineoplastic Agents/pharmacology , Calcium-Binding Proteins , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Female , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/immunology , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/immunology , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Membrane Proteins/immunology , Mice , Mice, SCID , Neoplasm Proteins/physiology , Ovarian Neoplasms/blood supply , Ovarian Neoplasms/metabolism , Receptors, Notch/physiology , Signal Transduction/drug effects , Species Specificity , Stromal Cells/drug effects , Stromal Cells/metabolism , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Xenograft Model Antitumor Assays
6.
PLoS One ; 9(11): e112371, 2014.
Article in English | MEDLINE | ID: mdl-25393540

ABSTRACT

BACKGROUND: The Notch ligand Delta-like 4 (Dll4) is highly expressed in vascular endothelium and has been shown to play a pivotal role in regulating tumor angiogenesis. Blockade of the Dll4-Notch pathway in preclinical cancer models has been associated with non-productive angiogenesis and reduced tumor growth. Given the cross-talk between the vascular endothelial growth factor (VEGF) and Delta-Notch pathways in tumor angiogenesis, we examined the activity of a function-blocking Dll4 antibody, REGN1035, alone and in combination with anti-VEGF therapy in renal cell carcinoma (RCC). METHODS AND RESULTS: Severe combined immunodeficiency (SCID) mice bearing patient-derived clear cell RCC xenografts were treated with REGN1035 and in combination with the multi-targeted tyrosine kinase inhibitor sunitinib or the VEGF blocker ziv-aflibercept. Immunohistochemical and immunofluorescent analyses were carried out, as well as magnetic resonance imaging (MRI) examinations pre and 24 hours and 2 weeks post treatment. Single agent treatment with REGN1035 resulted in significant tumor growth inhibition (36-62%) that was equivalent to or exceeded the single agent anti-tumor activity of the VEGF pathway inhibitors sunitinib (38-54%) and ziv-aflibercept (46%). Importantly, combination treatments with REGN1035 plus VEGF inhibitors resulted in enhanced anti-tumor effects (72-80% growth inhibition), including some tumor regression. Magnetic resonance imaging showed a marked decrease in tumor perfusion in all treatment groups. Interestingly, anti-tumor efficacy of the combination of REGN1035 and ziv-aflibercept was also observed in a sunitinib resistant ccRCC model. CONCLUSIONS: Overall, these findings demonstrate the potent anti-tumor activity of Dll4 blockade in RCC patient-derived tumors and a combination benefit for the simultaneous targeting of the Dll4 and VEGF signaling pathways, highlighting the therapeutic potential of this treatment modality in RCC.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antineoplastic Agents/chemistry , Carcinoma, Renal Cell/drug therapy , Intercellular Signaling Peptides and Proteins/chemistry , Kidney Neoplasms/drug therapy , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Adaptor Proteins, Signal Transducing , Animals , Antibodies, Monoclonal, Humanized , Calcium-Binding Proteins , Carcinoma, Renal Cell/metabolism , Cell Line, Tumor , Humans , Indoles/administration & dosage , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Kidney Neoplasms/metabolism , Male , Membrane Proteins/antagonists & inhibitors , Mice , Mice, SCID , Neovascularization, Pathologic , Pyrroles/administration & dosage , Receptors, Vascular Endothelial Growth Factor/administration & dosage , Recombinant Fusion Proteins/administration & dosage , Signal Transduction , Sunitinib , Vascular Endothelial Growth Factor A/metabolism , Xenograft Model Antitumor Assays
7.
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.

8.
Nat Med ; 18(1): 111-9, 2011 Dec 04.
Article in English | MEDLINE | ID: mdl-22138751

ABSTRACT

Mechanisms of epithelial cell renewal remain poorly understood in the mammalian kidney, particularly in the glomerulus, a site of cellular damage in chronic kidney disease. Within the glomerulus, podocytes--differentiated epithelial cells crucial for filtration--are thought to lack substantial capacity for regeneration. Here we show that podocytes rapidly lose differentiation markers and enter the cell cycle in adult mice in which the telomerase protein component TERT is conditionally expressed. Transgenic TERT expression in mice induces marked upregulation of Wnt signaling and disrupts glomerular structure, resulting in a collapsing glomerulopathy resembling those in human disease, including HIV-associated nephropathy (HIVAN). Human and mouse HIVAN kidneys show increased expression of TERT and activation of Wnt signaling, indicating that these are general features of collapsing glomerulopathies. Silencing transgenic TERT expression or inhibiting Wnt signaling through systemic expression of the Wnt inhibitor Dkk1 in either TERT transgenic mice or in a mouse model of HIVAN results in marked normalization of podocytes, including rapid cell-cycle exit, re-expression of differentiation markers and improved filtration barrier function. These data reveal an unexpected capacity of podocytes to reversibly enter the cell cycle, suggest that podocyte renewal may contribute to glomerular homeostasis and implicate the telomerase and Wnt-ß-catenin pathways in podocyte proliferation and disease.


Subject(s)
AIDS-Associated Nephropathy/metabolism , Kidney Glomerulus/metabolism , Kidney/metabolism , Podocytes/cytology , Telomerase/metabolism , Wnt Signaling Pathway , AIDS-Associated Nephropathy/genetics , Animals , Cell Differentiation , Cell Proliferation , Disease Models, Animal , Gene Expression Regulation , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Kidney/cytology , Kidney Glomerulus/cytology , Kidney Glomerulus/growth & development , Mice , Mice, Transgenic , Podocytes/metabolism , Telomerase/genetics
9.
Vasc Cell ; 3(1): 20, 2011 Sep 18.
Article in English | MEDLINE | ID: mdl-21923938

ABSTRACT

Tumor angiogenesis is an important target for cancer therapy, with most current therapies designed to block the VEGF signaling pathway. However, clinical resistance to anti-VEGF therapy highlights the need for targeting additional tumor angiogenesis signaling pathways. The endothelial Notch ligand Dll4 (delta-like 4) has recently emerged as a critical regulator of tumor angiogenesis and thus as a promising new therapeutic anti-angiogenesis target. Blockade of Dll4-Notch signaling in tumors results in excessive, non-productive angiogenesis with resultant inhibitory effects on tumor growth, even in some tumors that are resistant to anti-VEGF therapies. As Dll4 inhibitors are entering clinical cancer trials, this review aims to provide current perspectives on the function of the Dll4-Notch signaling axis during tumor angiogenesis and as a target for anti-angiogenic cancer therapy.

10.
Am J Pathol ; 178(2): 911-23, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21281822

ABSTRACT

Microvascular pericytes and perivascular fibroblasts have recently been identified as the source of scar-producing myofibroblasts that appear after injury of the kidney. We show that cross talk between pericytes and endothelial cells concomitantly dictates development of fibrosis and loss of microvasculature after injury. When either platelet-derived growth factor receptor (R)-ß signaling in pericytes or vascular endothelial growth factor (VEGF)R2 signaling in endothelial cells was blocked by circulating soluble receptor ectodomains, both fibrosis and capillary rarefaction were markedly attenuated during progressive kidney injury. Blockade of either receptor-mediated signaling pathway prevented pericyte differentiation and proliferation, but VEGFR2 blockade also attenuated recruitment of inflammatory macrophages throughout disease progression. Whereas injury down-regulated angiogenic VEGF164, the dys-angiogenic isomers VEGF120 and VEGF188 were up-regulated, suggesting that pericyte-myofibroblast differentiation triggers endothelial loss by a switch in secretion of VEGF isomers. These findings link fibrogenesis inextricably with microvascular rarefaction for the first time, add new significance to fibrogenesis, and identify novel therapeutic targets.


Subject(s)
Endothelium, Vascular/pathology , Kidney/blood supply , Kidney/pathology , Microvessels/pathology , Pericytes/metabolism , Signal Transduction , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Capillaries/metabolism , Capillaries/pathology , Cell Proliferation , Endothelium, Vascular/metabolism , Fibrosis , Humans , Immunity, Innate , Kidney/metabolism , Mice , Microvessels/metabolism , Neovascularization, Pathologic/complications , Neovascularization, Pathologic/metabolism , Pericytes/pathology , Protein Isoforms/metabolism , Receptor, Platelet-Derived Growth Factor beta/antagonists & inhibitors , Receptor, Platelet-Derived Growth Factor beta/metabolism , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors
11.
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
12.
Dev Biol ; 347(2): 325-36, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-20816797

ABSTRACT

Heart valve malformations are one of the most common types of birth defects, illustrating the complex nature of valve development. Vascular endothelial growth factor (VEGF) signaling is one pathway implicated in valve formation, however its specific spatial and temporal roles remain poorly defined. To decipher these contributions, we use two inducible dominant negative approaches in mice to disrupt VEGF signaling at different stages of embryogenesis. At an early step in valve development, VEGF signals are required for the full transformation of endocardial cells to mesenchymal cells (EMT) at the outflow tract (OFT) but not atrioventricular canal (AVC) endocardial cushions. This role likely involves signaling mediated by VEGF receptor 1 (VEGFR1), which is highly expressed in early cushion endocardium before becoming downregulated after EMT. In contrast, VEGFR2 does not exhibit robust cushion endocardium expression until after EMT is complete. At this point, VEGF signaling acts through VEGFR2 to direct the morphogenesis of the AVC cushions into mature, elongated valve leaflets. This latter role of VEGF requires the VEGF-modulating microRNA, miR-126. Thus, VEGF roles in the developing valves are dynamic, transitioning from a differentiation role directed by VEGFR1 in the OFT to a morphogenetic role through VEGFR2 primarily in the AVC-derived valves.


Subject(s)
Heart Valves/embryology , Vascular Endothelial Growth Factor A/physiology , Animals , Body Patterning/genetics , Body Patterning/physiology , Endocardium/embryology , Female , Gene Expression Regulation, Developmental , Mesoderm/embryology , Mice , Mice, Transgenic , MicroRNAs/genetics , Models, Cardiovascular , Pregnancy , Signal Transduction/genetics , Signal Transduction/physiology , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor Receptor-1/genetics , Vascular Endothelial Growth Factor Receptor-1/physiology , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/physiology
13.
Blood ; 115(23): 4631-3, 2010 Jun 10.
Article in English | MEDLINE | ID: mdl-20538815
14.
Arterioscler Thromb Vasc Biol ; 30(7): 1378-88, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20431070

ABSTRACT

OBJECTIVE: Vezf1 encodes an early zinc finger transcription factor that is essential for normal vascular development and functions in a dose-dependent manner. Here, we investigated the role of Vezf1 during processes of endothelial cell differentiation and maturation by studying mutant Vezf1 embryonic stem (ES) cells using the in vitro embryoid body differentiation model and the in vivo teratocarcinoma model. METHODS AND RESULTS: Vezf1-/- ES cell-derived embryoid bodies failed to form a well-organized vascular network and showed dramatic vascular sprouting defects. Our results indicate that the retinol pathway is an important mediator of Vezf1 function and that loss of Vezf1 results in reduced retinol/vitamin A signaling and aberrant extracellular matrix (ECM) formation. Unexpectedly, we also uncovered defects during in vitro differentiation of Vezf1-/- ES cells along hematopoietic cell lineages. Vezf1-/- ES cell-derived teratocarcinomas were able to spontaneously differentiate into cell types of all 3 germ layers. However, histological and immunohistochemical examination of these tumors showed decreased cell proliferation, delayed differentiation, and large foci of cells with extensive deposition of ECM. Embryoid bodies and teratocarcinomas derived from heterozygous ES cells displayed an intermediate phenotype. CONCLUSIONS: Together, these results suggest that Vezf1 is involved in early differentiation processes of the vasculature by regulating cell differentiation, proliferation, and ECM distribution and deposition.


Subject(s)
Cell Differentiation/genetics , Embryonic Stem Cells/metabolism , Endothelial Cells/metabolism , Gene Expression Regulation, Developmental , Kruppel-Like Transcription Factors/genetics , Mutation , Neovascularization, Physiologic/genetics , Animals , Cell Adhesion , Cell Proliferation , Cells, Cultured , DNA-Binding Proteins , Embryonal Carcinoma Stem Cells/metabolism , Embryonic Stem Cells/transplantation , Endothelial Cells/transplantation , Extracellular Matrix/metabolism , Genotype , Hematopoietic Stem Cells/metabolism , Kruppel-Like Transcription Factors/metabolism , Male , Mice , Mice, Nude , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Phenotype , Signal Transduction , Teratocarcinoma/blood supply , Teratocarcinoma/genetics , Teratocarcinoma/metabolism , Time Factors , Transcription Factors , Vitamin A/metabolism
15.
Am J Pathol ; 176(4): 2009-18, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20133818

ABSTRACT

Vascular endothelial growth factor (VEGF) is a key angiogenic factor in tumors, but less is known about what drives vascular remodeling in inflammation, where plasma leakage and leukocyte influx are prominent features. In chronic airway inflammation in mice infected by the bacterium Mycoplasma pulmonis (M. pulmonis), the segment of the microvasculature that supports leukocyte adhesion and migration expands through remodeling of capillaries into vessels with features of venules. Here, we report that the angiopoietin/Tie2 pathway is an essential driving force for capillary remodeling into venules in M. pulmonis-infected mouse airways. Similar to M. pulmonis infection, systemic overexpression of angiopoietin-1 (Ang1) resulted in remodeling of airway capillaries into venular-like vessels that expressed venous markers like P-selectin, ICAM-1, and EphB4 and were sites of leukocyte adhesion during lipopolysaccharide-induced acute inflammation. Ang1 and Ang2 protein increased in M. pulmonis-infected mouse airways but came from different cellular sources: Ang1 was expressed in infiltrating neutrophils and Ang2 in endothelial cells. Indeed, systemic administration of soluble Tie2 inhibited capillary remodeling, induction of venous markers, and leukocyte influx in M. pulmonis-infected mouse airways. Together, these findings suggest that blockade of the Ang/Tie2 pathway may represent a therapeutic approach in airway inflammation.


Subject(s)
Angiopoietin-1/metabolism , Capillaries/metabolism , Inflammation , Leukocytes/cytology , Receptor, TIE-2/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism , Adenoviridae/metabolism , Animals , Biological Transport , Intercellular Adhesion Molecule-1/biosynthesis , Mice , Mice, Inbred C57BL , Mycoplasma pulmonis/metabolism , Venules/metabolism
16.
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
17.
Proc Natl Acad Sci U S A ; 106(2): 641-6, 2009 Jan 13.
Article in English | MEDLINE | ID: mdl-19129494

ABSTRACT

Despite the importance of CNS blood vessels, the molecular mechanisms that regulate CNS angiogenesis and blood-brain barrier (BBB) formation are largely unknown. Here we analyze the role of Wnt/beta-catenin signaling in regulating the formation of CNS blood vessels. First, through the analysis of TOP-Gal Wnt reporter mice, we identify that canonical Wnt/beta-catenin signaling is specifically activated in CNS, but not non-CNS, blood vessels during development. This activation correlates with the expression of different Wnt ligands by neural progenitor cells in distinct locations throughout the CNS, including Wnt7a and Wnt7b in ventral regions and Wnt1, Wnt3, Wnt3a, and Wnt4 in dorsal regions. Blockade of Wnt/beta-catenin signaling in vivo specifically disrupts CNS, but not non-CNS, angiogenesis. These defects include reduction in vessel number, loss of capillary beds, and the formation of hemorrhagic vascular malformations that remain adherent to the meninges. Furthermore, we demonstrate that Wnt/beta-catenin signaling regulates the expression of the BBB-specific glucose transporter glut-1. Taken together these experiments reveal an essential role for Wnt/beta-catenin signaling in driving CNS-specific angiogenesis and provide molecular evidence that angiogenesis and BBB formation are in part linked.


Subject(s)
Central Nervous System/blood supply , Glucose Transporter Type 1/genetics , Neovascularization, Physiologic , Signal Transduction/physiology , Wnt Proteins/metabolism , beta Catenin/metabolism , Animals , Blood-Brain Barrier/chemistry , Gene Expression Regulation/physiology , Ligands , Mice , Neurons/chemistry , Stem Cells/chemistry , Wnt Proteins/analysis , beta Catenin/analysis
18.
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
19.
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
20.
Proc Natl Acad Sci U S A ; 105(29): 10185-90, 2008 Jul 22.
Article in English | MEDLINE | ID: mdl-18632559

ABSTRACT

The simultaneous targeting of both endothelial cells and pericytes via inhibition of VEGF receptor (VEGFR) and PDGFbeta receptor (PDGFRbeta) signaling, respectively, has been proposed to enhance the efficacy of antiangiogenic tumor therapy. Clinical and preclinical modeling of combined VEGFR and PDGFRbeta signaling inhibition, however, has used small molecule kinase inhibitors with inherently broad substrate specificities, precluding detailed examination of this hypothesis. Here, adenoviral expression of a soluble VEGFR2/Flk1 ectodomain (Ad Flk1-Fc) in combination with a soluble ectodomain of PDGFRbeta (Ad sPDGFRbeta) allowed highly selective inhibition of these pathways. The activity of Ad sPDGFRbeta was validated in vitro against PDGF-BB and in vivo with near-complete blockade of pericyte recruitment in the angiogenic corpus luteum, resulting in prominent hemorrhage, thus demonstrating an essential function for PDGF signaling during ovarian angiogenesis. Combination therapy with Ad PDGFRbeta and submaximal doses of Ad Flk1-Fc produced modest additive antitumor effects; however, no additivity was observed with maximal VEGF inhibition in numerous s.c. models. Notably, VEGF inhibition via Ad Flk1-Fc was sufficient to strongly suppress tumor endothelial and pericyte content as well as intratumoral PDGF-B mRNA, obscuring additive Ad sPDGFRbeta effects on pericytes or tumor volume. These studies using highly specific soluble receptors suggest that additivity between VEGFR and PDGFRbeta inhibition depends on the strength of VEGF blockade and appears minimal under conditions of maximal VEGF antagonism.


Subject(s)
Neovascularization, Pathologic , Neovascularization, Physiologic , Receptor, Platelet-Derived Growth Factor beta/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Adenoviridae/genetics , Animals , Corpus Luteum/blood supply , Corpus Luteum/cytology , Female , Genetic Therapy , Hemorrhage/etiology , Mice , Mice, Inbred C57BL , Neoplasms, Experimental/blood supply , Neoplasms, Experimental/genetics , Neoplasms, Experimental/therapy , Pericytes/cytology , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Receptor, Platelet-Derived Growth Factor beta/physiology , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction , Solubility , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...