Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Mol Ther ; 20(8): 1508-15, 2012 08.
Article in English | MEDLINE | ID: mdl-22828502

ABSTRACT

Selective inhibition of disease-related proteins underpins the majority of successful drug-target interactions. However, development of effective antagonists is often hampered by targets that are not druggable using conventional approaches. Here, we apply engineered zinc-finger protein transcription factors (ZFP TFs) to the endogenous phospholamban (PLN) gene, which encodes a well validated but recalcitrant drug target in heart failure. We show that potent repression of PLN expression can be achieved with specificity that approaches single-gene regulation. Moreover, ZFP-driven repression of PLN increases calcium reuptake kinetics and improves contractile function of cardiac muscle both in vitro and in an animal model of heart failure. These results support the development of the PLN repressor as therapy for heart failure, and provide evidence that delivery of engineered ZFP TFs to native organs can drive therapeutically relevant levels of gene repression in vivo. Given the adaptability of designed ZFPs for binding diverse DNA sequences and the ubiquity of potential targets (promoter proximal DNA), our findings suggest that engineered ZFP repressors represent a powerful tool for the therapeutic inhibition of disease-related genes, therefore, offering the potential for therapeutic intervention in heart failure and other poorly treated human diseases.


Subject(s)
Calcium-Binding Proteins/metabolism , Heart Failure/metabolism , Heart Failure/therapy , Transcription Factors/metabolism , Zinc Fingers/physiology , Adenoviridae/genetics , Animals , Blotting, Western , Calcium-Binding Proteins/genetics , Cell Line , Heart Failure/genetics , Humans , Kinetics , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/physiology , Rats , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/genetics , Zinc Fingers/genetics
2.
Mol Cell Biol ; 28(11): 3790-803, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18285456

ABSTRACT

Hypoxia-inducible transcription factor 1 (HIF-1) and HIF-2alpha regulate the expression of an expansive array of genes associated with cellular responses to hypoxia. Although HIF-regulated genes mediate crucial beneficial short-term biological adaptations, we hypothesized that chronic activation of the HIF pathway in cardiac muscle, as occurs in advanced ischemic heart disease, is detrimental. We generated mice with cardiac myocyte-specific deletion of the von Hippel-Lindau protein (VHL), an essential component of an E3 ubiquitin ligase responsible for suppressing HIF levels during normoxia. These mice were born at expected frequency and thrived until after 3 months postbirth, when they developed severe progressive heart failure and premature death. VHL-null hearts developed lipid accumulation, myofibril rarefaction, altered nuclear morphology, myocyte loss, and fibrosis, features seen for various forms of human heart failure. Further, nearly 50% of VHL(-/-) hearts developed malignant cardiac tumors with features of rhabdomyosarcoma and the capacity to metastasize. As compelling evidence for the mechanistic contribution of HIF-1alpha, the concomitant deletion of VHL and HIF-1alpha in the heart prevented this phenotype and restored normal longevity. These findings strongly suggest that chronic activation of the HIF pathway in ischemic hearts is maladaptive and contributes to cardiac degeneration and progression to heart failure.


Subject(s)
Heart Failure/genetics , Heart Failure/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia/genetics , Hypoxia/pathology , Myocardium/metabolism , Myocardium/pathology , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Animals , Capillaries/growth & development , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , ErbB Receptors/metabolism , Gene Deletion , Gene Transfer Techniques , Heart Failure/metabolism , Heart Neoplasms/genetics , Heart Neoplasms/metabolism , Heart Neoplasms/pathology , Hypoxia/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lipid Metabolism/genetics , Lipids/analysis , Mice , Mice, Knockout , Myocardium/chemistry , Neovascularization, Physiologic/genetics , Phosphorylation , Proto-Oncogene Proteins c-met/metabolism , ras Proteins/metabolism
3.
J Biol Chem ; 281(21): 14882-92, 2006 May 26.
Article in English | MEDLINE | ID: mdl-16556595

ABSTRACT

Among 10 adult mouse tissues tested, the p204 protein levels were highest in heart and skeletal muscle. We described previously that the MyoD-inducible p204 protein is required for the differentiation of cultured murine C2C12 skeletal muscle myoblasts to myotubes. Here we report that p204 was also required for the differentiation of cultured P19 murine embryonal carcinoma stem cells to beating cardiac myocytes. As shown by others, this process can be triggered by dimethyl sulfoxide (DMSO). We established that DMSO induced the formation of 204RNA and p204. Ectopic p204 could partially substitute for DMSO in inducing differentiation, whereas ectopic 204 antisense RNA inhibited the differentiation. Experiments with reporter constructs, including regulatory regions from the Ifi204 gene (encoding p204) in P19 cells and in cultured newborn rat cardiac myocytes, as well as chromatin coimmunoprecipitations with transcription factors, revealed that p204 expression was synergistically transactivated by the cardiac Gata4, Nkx2.5, and Tbx5 transcription factors. Furthermore, ectopic p204 triggered the expression of Gata4 and Nkx2.5 in P19 cells. p204 contains a nuclear export signal and was partially translocated to the cytoplasm during the differentiation. p204 from which the nuclear export signal was deleted was not translocated, and it did not induce differentiation. The various mechanisms by which p204 promoted the differentiation are reported in the accompanying article (Ding, B., Liu, C., Huang, Y., Yu, J., Kong, W., and Lengyel, P. (2006) J. Biol. Chem. 281, 14893-14906).


Subject(s)
GATA4 Transcription Factor/metabolism , Gene Expression Regulation, Developmental , Homeodomain Proteins/metabolism , Myocytes, Cardiac/metabolism , Nuclear Proteins/chemistry , Nuclear Proteins/physiology , Phosphoproteins/chemistry , Phosphoproteins/physiology , T-Box Domain Proteins/metabolism , Transcription Factors/metabolism , Amino Acid Sequence , Animals , Base Sequence , Cell Line, Tumor , Chromatin/metabolism , Homeobox Protein Nkx-2.5 , Mice , Molecular Sequence Data , Muscle, Skeletal/metabolism
4.
FASEB J ; 20(3): 479-81, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16423874

ABSTRACT

Advances in understanding the relationship between protein structure and DNA binding specificity have made it possible to engineer zinc finger protein (ZFP) transcription factors to specifically activate or repress virtually any gene. To evaluate the potential clinical utility of this approach for peripheral vascular disease, we investigated the ability of an engineered vascular endothelial growth factor (VEGFa)-activating ZFP (MVZ+426b) to induce angiogenesis and rescue hindlimb ischemia in a murine model. Hindlimb ischemia was surgically induced in advanced-age C57/BL6 mice. Adenovirus (Ad) encoding either MVZ+426b or the fluorescent marker dsRed was delivered to the adducter muscle of the ischemic hindlimb, and the effects on blood flow, limb salvage, and vascularization were assessed. Ad-MVZ+426b induced expression of VEGFa at the mRNA and protein levels and stimulated a significant increase in vessel counts in the ischemic limb. This was accompanied by significantly increased blood flow and limb salvage as measured serially for 4 wk. These data demonstrate that activation of the endogenous VEGFa gene by an engineered ZFP can induce angiogenesis in a clinically relevant model and further document the feasibility of designing ZFPs to therapeutically regulate gene expression in vivo.


Subject(s)
Gene Expression Regulation/genetics , Genetic Therapy , Genetic Vectors/therapeutic use , Ischemia/therapy , Neovascularization, Physiologic/genetics , Transcription Factors/physiology , Vascular Endothelial Growth Factor A/biosynthesis , Zinc Fingers/physiology , Adenoviridae/genetics , Aging , Amino Acid Sequence , Animals , Blood Flow Velocity , Feasibility Studies , Genes, Synthetic , Hindlimb/blood supply , Laser-Doppler Flowmetry , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Protein Engineering , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Recombinant Proteins , Structure-Activity Relationship , Transcription Factors/genetics , Vascular Endothelial Growth Factor A/genetics , Zinc Fingers/genetics
5.
Proc Natl Acad Sci U S A ; 102(1): 204-9, 2005 Jan 04.
Article in English | MEDLINE | ID: mdl-15615855

ABSTRACT

The functions of caveolae and/or caveolins in intact animals are beginning to be explored. Here, by using endothelial cell-specific transgenesis of the caveolin-1 (Cav-1) gene in mice, we show the critical role of Cav-1 in several postnatal vascular paradigms. First, increasing levels of Cav-1 do not increase caveolae number in the endothelium in vivo. Second, despite a lack of quantitative changes in organelle number, endothelial-specific expression of Cav-1 impairs endothelial nitric oxide synthase activation, endothelial barrier function, and angiogenic responses to exogenous VEGF and tissue ischemia. In addition, VEGF-mediated phosphorylation of Akt and its substrate, endothelial nitric oxide synthase, were significantly reduced in VEGF-treated Cav-1 transgenic mice, compared with WT littermates. The inhibitory effect of Cav-1 expression on the Akt-endothelial nitric oxide synthase pathway was specific because VEGF-stimulated phosphorylation of mitogen-activated protein kinase (ERK1/2) was elevated in the Cav-1 transgenics, compared with littermates. These data strongly support the idea that, in vivo, Cav-1 may modulate signaling pathways independent of its essential role in caveolae biogenesis.


Subject(s)
Caveolins/metabolism , Endothelium/metabolism , Neovascularization, Physiologic/physiology , Animals , Blood Vessels/metabolism , Caveolin 1 , Caveolins/genetics , Dogs , Mice , Mice, Transgenic , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/physiology , Vascular Endothelial Growth Factor Receptor-2/metabolism
6.
Circulation ; 110(21): 3300-5, 2004 Nov 23.
Article in English | MEDLINE | ID: mdl-15533866

ABSTRACT

BACKGROUND: After myocardial infarction (MI), bone marrow-derived cells (BMDCs) are found within the myocardium. The mechanisms determining BMDC recruitment to the heart remain unclear. We investigated the role of stromal cell-derived factor-1alpha (SDF-1) in this process. METHODS AND RESULTS: MI produced in mice by coronary ligation induced SDF-1 mRNA and protein expression in the infarct and border zone and decreased serum SDF-1 levels. By quantitative polymerase chain reaction, 48 hours after intravenous infusion of donor-lineage BMDCs, there were 80.5+/-15.6% more BDMCs in infarcted hearts compared with sham-operated controls (P<0.01). Administration of AMD3100, which specifically blocks binding of SDF-1 to its endogenous receptor CXCR4, diminished BMDC recruitment after MI by 64.2+/-5.5% (P<0.05), strongly suggesting a requirement for SDF-1 in BMDC recruitment to the infarcted heart. Forced expression of SDF-1 in the heart by adenoviral gene delivery 48 hours after MI doubled BMDC recruitment over MI alone (P<0.001) but did not enhance recruitment in the absence of MI, suggesting that SDF-1 can augment, but is not singularly sufficient for, BDMC recruitment to the heart. Gene expression analysis after MI revealed increased levels of several genes in addition to SDF-1, including those for vascular endothelial growth factor, matrix metalloproteinase-9, intercellular adhesion molecule-1, and vascular cell adhesion molecule-1, which might act in concert with SDF-1 to recruit BMDCs to the injured heart. CONCLUSIONS: SDF-1/CXCR4 interactions play a crucial role in the recruitment of BMDCs to the heart after MI and can further increase homing in the presence, but not in the absence, of injury.


Subject(s)
Bone Marrow Transplantation , Cell Movement/physiology , Chemokines, CXC/physiology , Myocardial Infarction/physiopathology , Receptors, CXCR4/physiology , Stem Cell Transplantation , Stem Cells/cytology , Animals , Benzylamines , Bone Marrow Cells/cytology , Cell Lineage , Chemokine CXCL12 , Chemokines, CXC/biosynthesis , Chemokines, CXC/blood , Chemokines, CXC/genetics , Cyclams , Female , Gene Expression Profiling , Gene Expression Regulation , Genetic Therapy , Heterocyclic Compounds/pharmacology , Intercellular Adhesion Molecule-1/biosynthesis , Intercellular Adhesion Molecule-1/genetics , Matrix Metalloproteinase 9/biosynthesis , Matrix Metalloproteinase 9/genetics , Mice , Mice, Inbred NOD , Mice, SCID , Myocardial Infarction/genetics , Myocardial Infarction/metabolism , Myocardial Infarction/therapy , Myocardium/pathology , Receptors, CXCR4/antagonists & inhibitors , Recombinant Fusion Proteins/physiology , Transduction, Genetic , Vascular Cell Adhesion Molecule-1/biosynthesis , Vascular Cell Adhesion Molecule-1/genetics , Vascular Endothelial Growth Factor A/biosynthesis , Vascular Endothelial Growth Factor A/genetics
7.
PLoS Biol ; 2(10): e288, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15328538

ABSTRACT

The physiological flux of oxygen is extreme in exercising skeletal muscle. Hypoxia is thus a critical parameter in muscle function, influencing production of ATP, utilization of energy-producing substrates, and manufacture of exhaustion-inducing metabolites. Glycolysis is the central source of anaerobic energy in animals, and this metabolic pathway is regulated under low-oxygen conditions by the transcription factor hypoxia-inducible factor 1alpha (HIF-1alpha). To determine the role of HIF-1alpha in regulating skeletal muscle function, we tissue-specifically deleted the gene encoding the factor in skeletal muscle. Significant exercise-induced changes in expression of genes are decreased or absent in the skeletal-muscle HIF-1alpha knockout mice (HIF-1alpha KOs); changes in activities of glycolytic enzymes are seen as well. There is an increase in activity of rate-limiting enzymes of the mitochondria in the muscles of HIF-1alpha KOs, indicating that the citric acid cycle and increased fatty acid oxidation may be compensating for decreased flow through the glycolytic pathway. This is corroborated by a finding of no significant decreases in muscle ATP, but significantly decreased amounts of lactate in the serum of exercising HIF-1alpha KOs. This metabolic shift away from glycolysis and toward oxidation has the consequence of increasing exercise times in the HIF-1alpha KOs. However, repeated exercise trials give rise to extensive muscle damage in HIF-1alpha KOs, ultimately resulting in greatly reduced exercise times relative to wild-type animals. The muscle damage seen is similar to that detected in humans in diseases caused by deficiencies in skeletal muscle glycogenolysis and glycolysis. Thus, these results demonstrate an important role for the HIF-1 pathway in the metabolic control of muscle function.


Subject(s)
Gene Expression Regulation , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Muscle, Skeletal/metabolism , Physical Conditioning, Animal/physiology , Adenosine Triphosphate/chemistry , Adenosine Triphosphate/metabolism , Alleles , Animals , Creatine Kinase/metabolism , Crosses, Genetic , Gene Deletion , Genotype , Glucose/metabolism , Glycogen/metabolism , Glycolysis , Hematocrit , Hemoglobins/metabolism , Hypoxia , Lactic Acid/blood , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron , Oxygen/metabolism , Physical Exertion , Polymerase Chain Reaction , Promoter Regions, Genetic , Reverse Transcriptase Polymerase Chain Reaction
8.
FASEB J ; 18(10): 1138-40, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15132980

ABSTRACT

At a resting pulse rate the heart consumes almost twice-as much oxygen per gram tissue as the brain and more than 43 times more than resting skeletal muscle (1). Unlike skeletal muscle, cardiac muscle cannot sustain anaerobic metabolism. Balancing oxygen demand with availability is crucial to cardiac function and survival, and regulated gene expression is a critical element of maintaining this balance. We investigated the role of the hypoxia-inducible transcription factor HIF-1alpha in maintaining this balance under normoxic conditions. Cardiac myocyte-specific HIF-1alpha gene deletion in the hearts of genetically engineered mice caused reductions in contractility, vascularization, high-energy phosphate content, and lactate production. This was accompanied by altered calcium flux and altered expression of genes involved in calcium handling, angiogenesis, and glucose metabolism. These findings support a central role for HIF-1alpha in coordinating energy availability and utilization in the heart and have implications for disease states in which cardiac oxygen delivery is impaired. Heart muscle requires a constant supply of oxygen. When oxygen supply does not match myocardial demand cardiac contractile dysfunction occurs, and prolongation of this mismatch leads to apoptosis and necrosis. Coordination of oxygen supply and myocardial demand involves immediate adaptations, such as coronary vasodilatation, and longer-term adaptations that include altered patterns of gene expression (2-4). How the expression of multiple genes is coordinated with oxygen availability in the heart and the impact of oxygen-dependent gene expression on cardiac function are insufficiently understood. Further elucidating these relationships may help clarify the molecular pathology of various cardiovascular disease states, including ischemic cardiomyopathy and myocardial hibernation (5, 6).


Subject(s)
Calcium Signaling/physiology , Coronary Circulation/physiology , DNA-Binding Proteins/physiology , Myocardial Contraction/physiology , Myocardium/metabolism , Myocytes, Cardiac/physiology , Nuclear Proteins/physiology , Transcription Factors/physiology , Animals , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Energy Metabolism , Gene Deletion , Gene Expression Regulation/physiology , Heart Function Tests , Hypoxia-Inducible Factor 1 , Hypoxia-Inducible Factor 1, alpha Subunit , Mice , Mice, Inbred C57BL , Mice, Knockout , Neovascularization, Physiologic/physiology , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Oxygen Consumption , RNA, Messenger/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/deficiency , Transcription Factors/genetics , Transcription, Genetic
9.
Am J Pathol ; 163(3): 869-78, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12937128

ABSTRACT

Decorin is a small proteoglycan that binds to transforming growth factor-beta (TGF-beta) and inhibits its activity. However, its interaction with platelet-derived growth factor (PDGF), involved in arterial repair after injury, is not well characterized. The objectives of this study were to assess decorin-PDGF and decorin-PDGF receptor (PDGFR) interactions, the in vitro effects of decorin on PDGF-stimulated smooth muscle cell (SMC) functions and the in vivo effects of decorin overexpression on arterial repair in a rabbit carotid balloon-injury model. Decorin binding to PDGF was demonstrated by solid-phase binding and affinity cross-linking assays. Decorin potently inhibited PDGF-stimulated PDGFR phosphorylation. Pretreatment of rabbit aortic SMC with decorin significantly inhibited PDGF-stimulated cell migration, proliferation, and collagen synthesis. Decorin overexpression by adenoviral-mediated gene transfection in balloon-injured carotid arteries significantly decreased intimal cross-sectional area and collagen content by approximately 50% at 10 weeks compared to beta-galactosidase-transfected or balloon-injured, non-transfected controls. This study shows that decorin binds to PDGF and inhibits its stimulatory activity on SMCs by preventing PDGFR phosphorylation. Decorin overexpression reduces intimal hyperplasia and collagen content after arterial injury. Decorin may be an effective therapy for the prevention of intimal hyperplasia after balloon angioplasty.


Subject(s)
Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Platelet-Derived Growth Factor/pharmacology , Proteoglycans/pharmacology , Angioplasty, Balloon/adverse effects , Animals , Apoptosis/drug effects , Cell Division/drug effects , Cell Movement/drug effects , Cells, Cultured , Collagen/biosynthesis , Collagen/metabolism , DNA/biosynthesis , Decorin , Extracellular Matrix Proteins , Fibronectins/metabolism , Hyperplasia/prevention & control , Muscle, Smooth, Vascular/physiology , Myocytes, Smooth Muscle/physiology , Necrosis , Phosphorylation/drug effects , Platelet-Derived Growth Factor/metabolism , Proteoglycans/genetics , Proteoglycans/metabolism , Rabbits , Receptor, Platelet-Derived Growth Factor beta/metabolism , Transgenes , Tunica Intima/pathology
10.
Nat Med ; 9(3): 357-62, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12598894

ABSTRACT

Small polybasic peptides derived from the transduction domains of certain proteins, such as the third alpha-helix of the Antennapedia (Antp) homeodomain, can cross the cell membrane through a receptor-independent mechanism. These cell-permeable molecules have been used as 'Trojan horses' to introduce biologically active cargo molecules such as DNA, peptides or proteins into cells. Using these cell-permeable peptides, we have developed an efficient and simple method to increase virally mediated gene delivery and protein expression in vitro and in vivo. Here, we show that cell-permeable peptides increase viral cell entry, improve gene expression at reduced titers of virus and improve efficacy of therapeutically relevant genes in vivo.


Subject(s)
Gene Transfer Techniques , Nuclear Proteins , Peptides/metabolism , Transcription Factors , Virus Replication/physiology , Adenoviridae/genetics , Adenoviridae/metabolism , Amino Acid Sequence , Animals , Antennapedia Homeodomain Protein , Arteries/cytology , Arteries/metabolism , COS Cells , Fluorescent Dyes/metabolism , Gene Expression Regulation , Genetic Therapy , Hindlimb/blood supply , Homeodomain Proteins/chemistry , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Ischemia , Molecular Sequence Data , Muscle, Skeletal/metabolism , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type III , Peptides/chemistry , Peptides/genetics , Recombinant Fusion Proteins/metabolism , Sequence Alignment
11.
Nat Med ; 8(12): 1427-32, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12415262

ABSTRACT

The relationship between the structure of zinc-finger protein (ZFP) transcription factors and DNA sequence binding specificity has been extensively studied. Advances in this field have made it possible to design ZFPs de novo that will bind to specific targeted DNA sequences. It has been proposed that such designed ZFPs may eventually be useful in gene therapy. A principal advantage of this approach is that activation of an endogenous gene ensures expression of the natural array of splice variants. Preliminary studies in tissue culture have validated the feasibility of this approach. The studies reported here were intended to test whether engineered transcription factors are effective in a whole-organism model. ZFPs were designed to regulate the endogenous gene encoding vascular endothelial growth factor-A (Vegfa). Expression of these new ZFPs in vivo led to induced expression of the protein VEGF-A, stimulation of angiogenesis and acceleration of experimental wound healing. In addition, the neovasculature resulting from ZFP-induced expression of Vegfa was not hyperpermeable as was that produced by expression of murine Vegfa(164) cDNA. These data establish, for the first time, that specifically designed transcription factors can regulate an endogenous gene in vivo and evoke a potentially therapeutic biophysiologic effect.


Subject(s)
Angiogenesis Inducing Agents , Gene Expression Regulation , Neovascularization, Physiologic , Protein Engineering/methods , Transcription Factors/physiology , Vascular Endothelial Growth Factor A , Zinc Fingers/physiology , 3T3 Cells , Amino Acid Sequence , Animals , Drug Design , Genetic Therapy , Mice , Models, Animal , Molecular Sequence Data , Recombinant Proteins , Transcription Factors/genetics
12.
Proc Natl Acad Sci U S A ; 99(11): 7734-9, 2002 May 28.
Article in English | MEDLINE | ID: mdl-12032352

ABSTRACT

Angiogenesis is regulated by means of a balance between activators and inhibitors. However, little is known regarding the regulation of the quiescent state of adult vessels. Corticotropin-releasing factor receptor 2 (CRFR2) is found in both endothelial and smooth muscle cells (SMCs) in the vasculature, where its function has remained elusive. We have investigated the role of CRFR2 as a determinant of tissue vascularization by comparing control and CRFR2-deficient mice with immunohistological and morphometric techniques. To define the mechanisms responsible for CRFR2 inhibition of angiogenesis, we have also examined in vitro the effect of ligand activation on cell proliferation, cell cycle protein phosphorylation, and capillary tube formation. Our results demonstrate that mice deficient for CRFR2 become hypervascularized postnatally. Activation of this receptor in vitro results in reduced vascular endothelial growth factor (VEGF) release from SMCs, an inhibition of SMC proliferation, and inhibition of capillary tube formation in collagen gels. Treatment of a subcutaneously injected gel matrix with a CRFR2 agonist inhibits growth factor-induced vascularization. Western blots show that cell cycle retinoblastoma protein, which is essential for cell cycle progression, is decreased by CRFR2 agonist treatment in SMCs. These results suggest that CRFR2 is a critical component of a pathway necessary for tonic inhibition of adult neovascularization. CRFR2 may be a potential target for therapeutic modulation of angiogenesis in cancer and ischemic cardiovascular disease.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Capillaries/physiology , Endothelium, Vascular/physiology , Muscle, Smooth, Vascular/physiology , Neovascularization, Physiologic/physiology , Receptors, Corticotropin-Releasing Hormone/physiology , Animals , Cell Division/physiology , Cells, Cultured , Endothelial Growth Factors/pharmacology , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Gene Expression Regulation/drug effects , Lymphokines/pharmacology , Mice , Mice, Knockout , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Neovascularization, Physiologic/drug effects , Receptors, Corticotropin-Releasing Hormone/deficiency , Receptors, Corticotropin-Releasing Hormone/genetics , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...