Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Mol Ther ; 15(1): 69-75, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17164777

ABSTRACT

Toxic neuropathy represents an important clinical problem in the use of the chemotherapeutic substances Taxol and thalidomide. Sensory neuropathy has a high incidence, lacks an effective treatment and is the dose-limiting factor for these drugs. The pathogenic basis of these neuropathies is unknown. We investigated the hypothesis that the experimental toxic neuropathies from Taxol and thalidomide results from destruction of vasa nervorum and can be reversed by the administration of an angiogenic cytokine. In animal models of Taxol- and thalidomide-induced neuropathy, nerve blood flow has been attenuated and the number of vasa nervorum has been reduced. Intramuscular gene transfer of naked plasmid DNA encoding VEGF-1 administered in parallel with Taxol injections completely inhibited deterioration of nerve function and diminution of the peripheral nerve vasculature. Gene therapy in animals with established Taxol- or thalidomide-induced neuropathies resulted in recovery of vascularity and improved nerve electrophysiology. These findings implicate microvascular damage as the basis for toxic neuropathy and suggest that angiogenic growth factors may constitute a novel treatment for this disorder.


Subject(s)
Neovascularization, Pathologic/drug therapy , Paclitaxel/pharmacology , Peripheral Nervous System Diseases/chemically induced , Thalidomide/pharmacology , Vasa Nervorum/drug effects , Vasa Nervorum/injuries , Vascular Endothelial Growth Factor A/pharmacology , Animals , Apoptosis/drug effects , Cells, Cultured , Drug Therapy, Combination , Electrophysiology , Endothelial Cells/drug effects , Endothelial Cells/enzymology , Humans , Injections, Intramuscular , Lectins/pharmacology , MAP Kinase Signaling System/drug effects , Male , Mitogen-Activated Protein Kinases/metabolism , Peripheral Nervous System Diseases/pathology , Phosphatidylinositol 3-Kinases/metabolism , Plasmids/genetics , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Renal Circulation
2.
J Clin Invest ; 111(5): 717-25, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12618526

ABSTRACT

Although lymphedema is a common clinical condition, treatment for this disabling condition remains limited and largely ineffective. Recently, it has been reported that overexpression of VEGF-C correlates with increased lymphatic vessel growth (lymphangiogenesis). However, the effect of VEGF-C-induced lymphangiogenesis on lymphedema has yet to be demonstrated. Here we investigated the impact of local transfer of naked plasmid DNA encoding human VEGF-C (phVEGF-C) on two animal models of lymphedema: one in the rabbit ear and the other in the mouse tail. In a rabbit model, following local phVEGF-C gene transfer, VEGFR-3 expression was significantly increased. This gene transfer led to a decrease in thickness and volume of lymphedema, improvement of lymphatic function demonstrated by serial lymphoscintigraphy, and finally, attenuation of the fibrofatty changes of the skin, the final consequences of lymphedema. The favorable effect of phVEGF-C on lymphedema was reconfirmed in a mouse tail model. Immunohistochemical analysis using lymphatic-specific markers: VEGFR-3, lymphatic endothelial hyaluronan receptor-1, together with the proliferation marker Ki-67 Ab revealed that phVEGF-C transfection potently induced new lymphatic vessel growth. This study, we believe for the first time, documents that gene transfer of phVEGF-C resolves lymphedema through direct augmentation of lymphangiogenesis. This novel therapeutic strategy may merit clinical investigation in patients with lymphedema.


Subject(s)
Endothelial Growth Factors/genetics , Genetic Therapy , Lymphatic System/physiology , Lymphedema/therapy , Neovascularization, Physiologic/drug effects , Amino Acid Sequence , Animals , Disease Models, Animal , Immunohistochemistry , Molecular Sequence Data , Phosphorylation , Rabbits , Reverse Transcriptase Polymerase Chain Reaction , Transgenes , Vascular Endothelial Growth Factor C , Vascular Endothelial Growth Factor Receptor-2/metabolism , Vascular Endothelial Growth Factor Receptor-3/genetics , Vascular Endothelial Growth Factor Receptor-3/metabolism
3.
J Mol Med (Berl) ; 84(6): 491-502, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16389546

ABSTRACT

Fibroblast growth factor 1 (FGF1) is an angiogenic factor known to play a role in the growth of arteries. The purpose of this study was to evaluate the usefulness of direct intramuscular injection of an optimized expression plasmid encoding FGF1 to augment collateral formation and tissue perfusion in a rabbit ischemic hindlimb model. Truncated FGF1 fused to the human fibroblast interferon (FIN) signal peptide was expressed from a newly designed plasmid backbone with an improved safety profile for gene therapy applications. In vitro, optimization of plasmid design yielded in a dramatic increase in expression efficiency for FGF1, independent of the presence of a signal peptide, as analyzed by Western Blotting. In vivo, successful transgene expression could be demonstrated by FGF1 immunostaining after gene application. FGF1 plasmid containing FIN signal peptide (100, 500, and 1,000 mug), when injected into ischemic muscle areas of rabbits 10 days after ligation of the external iliac artery, exhibited a pronounced therapeutic effect on collateral formation to the ischemic hindlimb in a dose-depending manner, as assessed by physiological (blood pressure ratio, maximal intra-arterial Doppler flow) and anatomical (angiographic score, histologic evaluation of capillary density) measurements 30 days after therapy, compared to saline or lacZ control plasmid. FGF1 plasmid without a signal peptide sequence resulted in a comparable therapeutic effect on collateral formation at comparable doses (500 and 1,000 mug). Our results indicate that intramuscular FGF1 gene application could be useful to stimulate collateral formation in a situation of chronic peripheral ischemia. The presence of a signal peptide does not seem to be obligatory to achieve bioactivity of intramuscular transfected FGF1. An optimized vector design improved both biosafety of gene transfer and expression efficiency of the transgene, rendering this vector highly suitable for human gene therapy. Therefore, this new generation vector encoding FGF1 might be useful as an alternative treatment for patients with chronic ischemic disorders not amenable to conventional therapy.


Subject(s)
Fibroblast Growth Factor 1/genetics , Gene Transfer Techniques , Hindlimb/blood supply , Ischemia/therapy , Muscle, Skeletal/blood supply , Neovascularization, Physiologic , Amino Acid Sequence , Animals , Blood Pressure , Cells, Cultured , Fibroblast Growth Factor 1/biosynthesis , Genetic Vectors , Humans , Injections, Intramuscular , Interferon-beta/genetics , Interferon-beta/physiology , Male , Molecular Sequence Data , Muscle, Skeletal/metabolism , Plasmids , Protein Sorting Signals/genetics , Protein Sorting Signals/physiology , Rabbits
4.
Circulation ; 111(17): 2210-8, 2005 May 03.
Article in English | MEDLINE | ID: mdl-15851594

ABSTRACT

BACKGROUND: Ephrin-B2, one of the transmembrane ligands, is a genetic marker of arterial endothelial cells (ECs) at embryonic stages and is essential for cardiovascular development, but its roles in ischemic cardiovascular disease are not well understood. In this study, we focused on the function of ephrin-B2 in postnatal neovascularization. METHODS AND RESULTS: We found that ephrin-B2 is exclusively expressed and significantly upregulated in the arterial vasculature after the initial angiogenic responses in tissue ischemia. Upregulation of ephrin-B2 is also observed in EC cordlike formation in vitro. Interestingly, ephrin-B2 expression on ECs was enhanced by promotive angiogenic growth factors, such as vascular endothelial growth factor, basic fibroblast growth factor, and hepatocyte growth factor, whereas it was attenuated by angiopoietin-1, a factor for blood vessel maturation. Moreover, an ephrin-B2-rich environment was shown to induce neovascularization mainly through venous angiogenesis in an in vivo cornea micropocket assay. CONCLUSIONS: Our study indicates that the ephrin-B2 ligand is likely to have functional expression on angiogenic arterial ECs and induce a subsequent promotive effect on venous vessels during postnatal neovascularization.


Subject(s)
Ephrin-B2/genetics , Ephrin-B2/physiology , Neovascularization, Physiologic , Up-Regulation/drug effects , Animals , Arteries , Cells, Cultured , Disease Models, Animal , Endothelium, Vascular/cytology , Endothelium, Vascular/physiology , Growth Substances/pharmacology , Hindlimb/blood supply , Humans , Ischemia , Limbus Corneae/blood supply , Mice , Mice, Transgenic , Veins
5.
Circulation ; 111(16): 2073-85, 2005 Apr 26.
Article in English | MEDLINE | ID: mdl-15851615

ABSTRACT

BACKGROUND: Diabetic cardiomyopathy (DCM) is characterized by microvascular pathology and interstitial fibrosis, which leads to progressive heart failure; however, the pathogenesis of DCM remains uncertain. METHODS AND RESULTS: Using the streptozotocin-induced diabetic rat model, we evaluated the natural course of DCM over a period of 1 year by serial echocardiography, Western blot analysis for vascular endothelial growth factor (VEGF), endothelial progenitor cell assays, myocardial blood flow measurements, and histopathologic analysis that included terminal dUTP nick end-labeling (TUNEL), capillary and cardiomyocyte density, and fibrosis area. Downregulation of myocardial VEGF expression preceded all other features of DCM and was followed by increased apoptosis of endothelial cells, decreased numbers of circulating endothelial progenitor cells, decreased capillary density, and impaired myocardial perfusion. Apoptosis and necrosis of cardiomyocytes ensued, along with fibrosis and progressive diastolic and then systolic dysfunction. To provide further evidence of the central role of VEGF in the pathophysiology of DCM, we replenished myocardial VEGF expression using naked DNA gene therapy via direct intramyocardial injection of plasmid DNA encoding VEGF (phVEGF165). VEGF-replenished rats showed increased capillary density, decreased endothelial cell and cardiomyocyte apoptosis, and in situ differentiation of bone marrow-derived endothelial progenitor cells into endothelial cells. These anatomic findings were accompanied by significant improvements in cardiac function. CONCLUSIONS: These findings suggest that downregulation of VEGF may compromise microvascular homeostasis in the myocardium and thereby play a central role in the pathogenesis of DCM.


Subject(s)
Cardiomyopathies/etiology , Diabetes Complications/etiology , Myocardium/chemistry , Vascular Endothelial Growth Factor A/administration & dosage , Vascular Endothelial Growth Factor A/analysis , Animals , Cardiomyopathies/therapy , Diabetes Complications/therapy , Diabetes Mellitus, Experimental , Down-Regulation , Female , Genetic Therapy , Homeostasis/drug effects , Male , Microcirculation/drug effects , Microcirculation/physiopathology , Rats , Rats, Inbred F344 , Rats, Sprague-Dawley , Treatment Outcome , Vascular Endothelial Growth Factor A/genetics
6.
Circulation ; 111(20): 2662-70, 2005 May 24.
Article in English | MEDLINE | ID: mdl-15897348

ABSTRACT

BACKGROUND: Toxic neuropathies induced by cisplatin and other chemotherapeutic agents are important clinical problems because of their high incidence, their lack of effective treatment, and the fact that neuropathy represents a dose-limiting factor for these therapies. The pathogenic basis for toxic neuropathies induced by chemotherapeutic agents has not been completely elucidated. METHODS AND RESULTS: We investigated the hypothesis that experimental toxic neuropathy results from an antiangiogenic effect of these drugs, resulting in destruction of the vasa nervorum, and accordingly that the neuropathy could be prevented or reversed by locally administered VEGF gene transfer without augmenting tumor growth. In an animal model of cisplatin-induced neuropathy, nerve blood flow was markedly attenuated, and there was a profound reduction in the number of vasa nervorum associated with marked endothelial cell apoptosis, resulting in a severe peripheral neuropathy with focal axonal degeneration characteristic of ischemic neuropathy. After intramuscular gene transfer of naked plasmid DNA encoding VEGF-1 in animals with an established neuropathy, vascularity and blood flow returned to levels similar to those of control rats, peripheral nerve function was restored, and histological nerve architecture was normalized. Gene therapy administered in parallel with cisplatin chemotherapy completely attenuated endothelial cell apoptosis and inhibited destruction of nerve vasculature, deterioration of nerve function, and axonal degeneration. In a rat tumor model, VEGF gene transfer administered locally did not alter tumor growth or vascularity. CONCLUSIONS: These findings implicate microvascular damage as the basis for toxic neuropathy induced by cisplatin and suggest that local angiogenic gene therapy may constitute a novel prevention or treatment for this disorder without augmenting tumor growth or vascularization.


Subject(s)
Cisplatin/adverse effects , Peripheral Nervous System Diseases/prevention & control , Vascular Endothelial Growth Factor A/administration & dosage , Angiogenesis Inhibitors/adverse effects , Animals , Apoptosis , Endothelium, Vascular/cytology , Genetic Therapy , Humans , Male , Neoplasms/blood supply , Neoplasms/complications , Neoplasms/drug therapy , Peripheral Nervous System Diseases/chemically induced , Peripheral Nervous System Diseases/therapy , Rats , Rats, Sprague-Dawley , Tumor Burden/drug effects , Umbilical Veins/cytology , Vasa Nervorum/drug effects , Vascular Endothelial Growth Factor A/therapeutic use
7.
Circ Res ; 93(2): 162-9, 2003 Jul 25.
Article in English | MEDLINE | ID: mdl-12829616

ABSTRACT

Tumor necrosis factor-alpha (TNF-alpha) is expressed locally in the vessel wall after angioplasty and induces growth arrest and apoptosis in endothelial cells (ECs), thereby delaying reendothelialization. Prior studies have shown that direct antagonism of TNF-alpha, using a systemically administered soluble receptor, can enhance endothelial recovery and reduce neointimal thickening. These studies have also shown that downregulation of the transcription factor E2F1 was a key mechanism of TNF's effect on ECs. We now show that Ad-E2F1 overexpression at sites of balloon injury accelerates functional endothelial recovery, consistent with the prior in vitro findings. Moreover these studies also reveal divergent effects of TNF-alpha and overexpression of E2F1 on ECs versus VSMCs. TNF-alpha exposure of VSMCs had no affect on proliferation or apoptosis, in contrast to the effect seen in ECs. In Ad-E2F1-transduced VSMCs, however, TNF-alpha-induced marked apoptosis in contrast to the survival effect seen in ECs. Finally, these studies suggest that differential activation of NF-kappaB may play a key role in mediating these opposing effects. Nuclear translocation and transcriptional activity of NF-kappaB was markedly attenuated in Ad-E2F1-transduced VSMCs, whereas it remained active in similarly treated ECs when the cells were exposed to TNF-alpha. These studies reveal that overexpression of Ad-E2F1 primes VSMCs to TNF-alpha-induced apoptosis. Furthermore, E2F1 potentiates VSMC death by blocking antiapoptotic signaling pathway through inhibition of NF-kappaB activation. The divergent responses of VSMCs and ECs to E2F1 overexpression provide unique therapeutic possibilities: simultaneously targeting the cell cycle of two different cell types, within same tissue microenvironment resulting in opposite and biologically complimentary effects.


Subject(s)
Carotid Artery Injuries/metabolism , Cell Cycle Proteins , DNA-Binding Proteins , Endothelium, Vascular/metabolism , Muscle, Smooth, Vascular/metabolism , Transcription Factors/metabolism , Tunica Intima/growth & development , Active Transport, Cell Nucleus , Animals , Apoptosis , Carotid Artery Injuries/pathology , Caspases/metabolism , Cattle , Cell Division/drug effects , Cell Division/genetics , Cells, Cultured , Disease Models, Animal , E2F Transcription Factors , E2F1 Transcription Factor , Endothelium, Vascular/cytology , Gene Expression Regulation , Genes, Reporter , Humans , Hyperplasia/pathology , I-kappa B Proteins/metabolism , Mice , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/pathology , NF-KappaB Inhibitor alpha , NF-kappa B/genetics , NF-kappa B/metabolism , Rats , Rats, Sprague-Dawley , Recovery of Function , Transcription Factors/genetics , Transfection , Tumor Necrosis Factor-alpha/pharmacology , Tunica Intima/injuries , Tunica Intima/pathology
8.
Circulation ; 106(9): 1133-9, 2002 Aug 27.
Article in English | MEDLINE | ID: mdl-12196341

ABSTRACT

BACKGROUND: The regulatory molecule for cell life span, telomerase, was modified by human telomerase reverse transcriptase (hTERT) gene transfer to investigate its effect on regenerative properties of endothelial progenitor cells (EPCs) in neovascularization. METHODS AND RESULTS: Telomerase activity was enhanced in hTERT-transduced EPCs (Td-TERTs) (1.2-fold versus no transduced EPCs [no-Td] and 1.2-fold versus GFP-transduced EPCs [Td/GFPs] at day 8; 5.2-fold versus no-Td and 4.8-fold versus Td/GFP at day 21, respectively) Mitogenic capacity in Td/TERTs exceeded that in Td/GFPs at day 8 (0.62+/-0.02 versus 0.53+/-0.01, respectively; P<0.01). Vascular endothelial growth factor-induced cell migration in EPCs was markedly enhanced by hTERT overexpression (Td/TERTs versus Td/GFPs, 292+/-12 versus 174+/-6 cells, respectively; P<0.01). hTERT overexpression has rescued EPCs from starvation-induced cell apoptosis, an outcome that was further enhanced in response to vascular endothelial growth factor. The colony appearance of totally differentiated endothelial cells (tdECs) was detected before day 30 only in Td/TERT, whereas no tdEC colonies could be detected in both Td/GFPs and no-Tds. Finally, we investigated in vivo transplantation of heterologous EPCs. Td/TERTs dramatically improved postnatal neovascularization in terms of limb salvage by 4-fold in comparison with that of Td/GFPs; limb perfusion was measured by laser Doppler (0.77+/-0.10 versus 0.47+/-0.06; P=0.02), and capillary density (224+/-78 versus 90+/-40 capillaries/mm2; P<0.01). CONCLUSIONS: These findings provide the novel evidence that telomerase activity contributes to EPC angiogenic properties; mitogenic activity, migratory activity, and cell survival. This enhanced regenerative activity of EPCs by hTERT transfer will provide novel therapeutical strategy for postnatal neovascularization in severe ischemic disease patients.


Subject(s)
Endothelium, Vascular/metabolism , Regeneration , Stem Cells/metabolism , Telomerase/biosynthesis , Telomerase/metabolism , Adenoviridae/genetics , Animals , Apoptosis , Cell Differentiation , Cell Division , Cell Movement , Cells, Cultured , Cellular Senescence/physiology , DNA-Binding Proteins , Disease Models, Animal , Endothelium, Vascular/cytology , Enzyme Activation , Gene Expression , Hindlimb/blood supply , Hindlimb/physiopathology , Humans , Ischemia/physiopathology , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/metabolism , Mice , Mice, Nude , Regeneration/physiology , Reverse Transcriptase Polymerase Chain Reaction , Stem Cells/cytology , Telomerase/genetics , Transduction, Genetic , beta-Galactosidase/metabolism
9.
Circulation ; 107(9): 1322-8, 2003 Mar 11.
Article in English | MEDLINE | ID: mdl-12628955

ABSTRACT

BACKGROUND: Stromal cell-derived factor-1 (SDF-1) is a chemokine considered to play an important role in the trafficking of hematopoietic stem cells. Given the close relationship between hematopoietic stem cells and endothelial progenitor cells (EPCs), we investigated the effect of SDF-1 on EPC-mediated vasculogenesis. METHODS AND RESULTS: Flow cytometric analysis demonstrated expression of CXCR4, the receptor of SDF-1, by 66+/-3% of EPCs after 7 days in culture. In vitro modified Boyden chamber assay showed a dose-dependent EPC migration toward SDF-1 (control versus 10 ng/mL SDF-1 versus 100 ng/mL SDF-1, 24+/-2 versus 71+/-3 versus 140+/-6 cells/mm2; P<0.0001). SDF-1 attenuated EPC apoptosis (control versus SDF-1, 27+/-1 versus 7+/-1%; P<0.0001). To investigate the effect of SDF-1 in vivo, we locally injected SDF-1 into athymic ischemic hindlimb muscle of nude mice combined with human EPC transplantation to determine whether SDF-1 augmented EPC-induced vasculogenesis. Fluorescence microscopic examination disclosed increased local accumulation of fluorescence-labeled EPCs in ischemic muscle in the SDF-1 treatment group (control versus SDF-1=241+/-25 versus 445+/-24 cells/mm2, P<0.0001). At day 28 after treatment, ischemic tissue perfusion was improved in the SDF-1 group and capillary density was also increased. (control versus SDF-1, 355+/-26 versus 551+/-30 cells/mm2; P<0.0001). CONCLUSION: These findings indicate that locally delivered SDF-1 augments vasculogenesis and subsequently contributes to ischemic neovascularization in vivo by augmenting EPC recruitment in ischemic tissues.


Subject(s)
Chemokines, CXC/pharmacology , Endothelium, Vascular/cytology , Hematopoietic Stem Cells/physiology , Ischemia/therapy , Neovascularization, Physiologic , Animals , Apoptosis , Capillaries/anatomy & histology , Capillaries/growth & development , Cell Movement , Cell Separation , Cells, Cultured , Chemokine CXCL12 , Endothelial Growth Factors/biosynthesis , Endothelial Growth Factors/genetics , Flow Cytometry , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/drug effects , Hindlimb/blood supply , Humans , Intercellular Signaling Peptides and Proteins/biosynthesis , Intercellular Signaling Peptides and Proteins/genetics , Ischemia/metabolism , Ischemia/physiopathology , Lymphokines/biosynthesis , Lymphokines/genetics , Male , Mice , Mice, Nude , Muscle, Skeletal/blood supply , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , RNA, Messenger/biosynthesis , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
10.
Circulation ; 108(4): 479-85, 2003 Jul 29.
Article in English | MEDLINE | ID: mdl-12860919

ABSTRACT

BACKGROUND: Hedgehog (Hh) proteins are morphogens regulating epithelial-mesenchymal signaling during several crucial processes of embryonic development, including muscle patterning. Sonic (Shh), Indian (Ihh), and Desert (Dhh) hedgehog constitute the repertoire of Hh genes in humans. The activities of all 3 are transduced via the Patched (Ptc1) receptor. Recent observations indicate that exogenous administration of Shh induces angiogenesis. Here, we studied whether the endogenous Hh pathway, in addition to its functions during embryogenesis, plays a physiological role in muscle regeneration after ischemia in adults. METHODS AND RESULTS: We found that skeletal muscle ischemia induces strong local upregulation of Shh mRNA and protein. In addition, the Ptc1 receptor is activated in interstitial mesenchymal cells within the ischemic area, indicating that these cells respond to Shh and that the Shh pathway is functional. We also found that Shh-responding cells produce vascular endothelial growth factor under ischemic conditions and that systemic treatment with a Shh-blocking antibody inhibits the local angiogenic response and the upregulation of vascular endothelial growth factor. CONCLUSIONS: Our study shows that the Hh signaling may be recapitulated postnatally in adult and fully differentiated muscular tissues and has a regulatory role on angiogenesis during muscle regeneration after ischemia. These findings demonstrate a novel biological activity for the Hh pathway with both fundamental and potential therapeutic implications.


Subject(s)
Ischemia/physiopathology , Muscle, Skeletal/physiopathology , Signal Transduction/physiology , Trans-Activators/physiology , Animals , Antibodies, Blocking/pharmacology , Blotting, Western , Disease Models, Animal , Endothelial Growth Factors/biosynthesis , Female , Hedgehog Proteins , Hindlimb/blood supply , Hindlimb/physiopathology , Immunohistochemistry , Intercellular Signaling Peptides and Proteins/biosynthesis , Intracellular Signaling Peptides and Proteins , Ischemia/pathology , Lymphokines/biosynthesis , Male , Membrane Proteins/metabolism , Mesoderm/metabolism , Mesoderm/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Muscle, Skeletal/blood supply , Muscle, Skeletal/pathology , Neovascularization, Physiologic/drug effects , Neovascularization, Physiologic/genetics , Patched Receptors , Patched-1 Receptor , RNA, Messenger/metabolism , Receptors, Cell Surface , Trans-Activators/antagonists & inhibitors , Trans-Activators/genetics , Up-Regulation/drug effects , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
11.
Circulation ; 105(6): 732-8, 2002 Feb 12.
Article in English | MEDLINE | ID: mdl-11839630

ABSTRACT

BACKGROUND: Previous studies have established that bone marrow-derived endothelial progenitor cells (EPCs) are present in the systemic circulation. In the current study, we investigated the hypothesis that gene transfer can be used to achieve phenotypic modulation of EPCs. METHODS AND RESULTS: In vitro, ex vivo murine vascular endothelial growth factor (VEGF) 164 gene transfer augmented EPC proliferative activity and enhanced adhesion and incorporation of EPCs into quiescent as well as activated endothelial cell monolayers. To determine if such phenotypic modulation may facilitate therapeutic neovascularization, heterologous EPCs transduced with adenovirus encoding VEGF were administered to athymic nude mice with hindlimb ischemia; neovascularization and blood flow recovery were both improved, and limb necrosis/autoamputation were reduced by 63.7% in comparison with control animals. The dose of EPCs used for the in vivo experiments was 30 times less than that required in previous trials of EPC transplantation to improve ischemic limb salvage. Necropsy analysis of animals that received DiI-labeled VEGF-transduced EPCs confirmed that enhanced EPC incorporation demonstrated in vitro contributed to in vivo neovascularization as well. CONCLUSIONS: In vitro, VEGF EPC gene transfer enhances EPC proliferation, adhesion, and incorporation into endothelial cell monolayers. In vivo, gene-modified EPCs facilitate the strategy of cell transplantation to augment naturally impaired neovascularization in an animal model of experimentally induced limb ischemia.


Subject(s)
Endothelial Growth Factors/pharmacology , Endothelium, Vascular/drug effects , Genetic Therapy/methods , Ischemia/drug therapy , Lymphokines/pharmacology , Regeneration/drug effects , Stem Cells/metabolism , Adenoviridae/genetics , Animals , Cell Count , Cell Division/drug effects , Cell Division/physiology , Cells, Cultured , Dendritic Cells , Disease Models, Animal , Endothelial Growth Factors/genetics , Endothelial Growth Factors/metabolism , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Female , Gene Transfer, Horizontal , Genetic Vectors/genetics , Genetic Vectors/metabolism , Genetic Vectors/pharmacology , Humans , Ischemia/pathology , Ischemia/physiopathology , Lymphokines/genetics , Lymphokines/metabolism , Mice , Mice, Nude , Microcirculation/drug effects , Microcirculation/metabolism , Microcirculation/pathology , Muscle, Skeletal/blood supply , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Neovascularization, Physiologic/drug effects , Stem Cell Transplantation , Stem Cells/cytology , Stem Cells/drug effects , Transgenes , Treatment Outcome , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
12.
Circulation ; 105(25): 3017-24, 2002 Jun 25.
Article in English | MEDLINE | ID: mdl-12081997

ABSTRACT

BACKGROUND: Primary and secondary prevention trials suggest that statins possess favorable effects independent of cholesterol reduction. We investigated whether statin therapy may also accelerate reendothelialization after carotid balloon injury. METHODS AND RESULTS: Simvastatin treatment in 34 male Sprague-Dawley rats accelerated reendothelialization of the balloon-injured arterial segments (reendothelialized area at 2 weeks, 12.3+/-1.8 versus 5.4+/-1.1 mm2, P< 0.01) and resulted in a dose-dependent (0.2 or 1 mg/kg IP) significant reduction in neointimal thickening at 2, 3, and 4 weeks compared with saline-injected controls (n=18). To elucidate the mechanism, we investigated the contribution of bone marrow-derived endothelial progenitor cells (EPCs) by bone marrow transplantation from Tie2/lacZ mice to background mice or nude rats. X-gal staining of mouse carotid artery specimens revealed a 2.9-fold increase in the number of beta-gal-positive cells per square millimeter appearing on the carotid artery luminal surface at 2 weeks, and double-fluorescence immunohistochemistry disclosed a significant 5-fold increase in the number of double-positive cells (beta-gal, isolectin B4) on the luminal surface in carotid arteries of statin-treated nude rats (20+/-3 versus 4+/-1 cells/mm surface length, P<0.005). Statins increased circulating rat EPCs (2.4-fold at 2 weeks and 2.5-fold at 4 weeks, P<0.001) and induced adhesiveness of cultured human EPCs by upregulation of the integrin subunits alpha5, beta1, alpha(v), and beta5 of human EPCs as shown by reverse transcription-polymerase chain reaction and fluorescence-activated cell sorting. CONCLUSIONS: These findings establish additional mechanisms by which statins may specifically preempt disordered vascular wall pathology and constitute physiological evidence that EPC mobilization represents a functionally relevant consequence of statin therapy.


Subject(s)
Arterial Occlusive Diseases/drug therapy , Endothelium, Vascular/growth & development , Hematopoietic Stem Cells/drug effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Simvastatin/pharmacology , Angioplasty, Balloon/adverse effects , Animals , Arterial Occlusive Diseases/etiology , Arterial Occlusive Diseases/pathology , Bone Marrow Transplantation , Carotid Stenosis/drug therapy , Carotid Stenosis/etiology , Carotid Stenosis/pathology , Cell Adhesion/drug effects , Cell Division/drug effects , Cell Movement/drug effects , Cells, Cultured , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/physiology , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Integrins/metabolism , Kinetics , Male , Mice , Rats , Rats, Sprague-Dawley , Simvastatin/therapeutic use
13.
Circulation ; 108(25): 3115-21, 2003 Dec 23.
Article in English | MEDLINE | ID: mdl-14676142

ABSTRACT

BACKGROUND: We hypothesized that estrogen-induced acceleration of reendothelialization might be mediated in part by effects involving mobilization and incorporation of bone marrow-derived endothelial progenitor cells (EPCs). METHODS AND RESULTS: Carotid injury was induced in ovariectomized wild-type mice receiving either 17beta-estradiol or placebo. Estradiol treatment significantly accelerated reendothelialization of injured arterial segments within 7 days and resulted in a significant reduction of medial thickness 14 and 21 days after the injury. Significant increases in circulating EPCs 3 days after the injury were observed in the estradiol group compared with placebo-treated mice. These data were further supported by fluorescence-activated cell sorting analysis, which disclosed a significant increase in Sca-1/Flk-1-positive cells in estradiol versus control mice. To evaluate the effects of estradiol on bone marrow-derived EPC incorporation at sites of reendothelialization, carotid injury was established in ovariectomized wild-type mice transplanted with bone marrow from transgenic donors expressing beta-galactosidase transcriptionally regulated by the Tie-2 promoter. Significantly greater numbers of X-gal-positive cells were observed at reendothelialized areas in the estradiol group 3 days after injury as compared with placebo. Fluorescent immunohistochemistry 14 days after the injury documented a marked increase in cells expressing both beta-gal, indicating bone marrow origin and Tie-2 expression, and isolectin B4, also indicating endothelial lineage, in the estradiol group compared with control. In contrast, estradiol did not accelerate reendothelialization or augment EPC mobilization into the peripheral circulation after injury in endothelial nitric oxide synthase-deficient mice (eNOS-/-). Furthermore, estradiol exhibited direct stimulatory effects on EPC mitogenic and migration activity and inhibited EPC apoptosis. CONCLUSIONS: Estradiol accelerates reendothelialization and attenuates medial thickening after carotid injury in part by augmenting mobilization and proliferation of bone marrow-derived EPCs and their incorporation into the recovering endothelium at the site of injury.


Subject(s)
Arterial Occlusive Diseases/drug therapy , Bone Marrow Cells/physiology , Endothelium, Vascular/cytology , Estradiol/therapeutic use , Nitric Oxide Synthase/physiology , Stem Cells/physiology , Animals , Apoptosis/drug effects , Bone Marrow Cells/drug effects , Carotid Arteries/cytology , Carotid Arteries/pathology , Carotid Stenosis/drug therapy , Carotid Stenosis/etiology , Carotid Stenosis/pathology , Cell Division/drug effects , Cell Movement/drug effects , Cells, Cultured , Endothelium, Vascular/pathology , Endothelium, Vascular/physiology , Estradiol/pharmacology , Female , Kinetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type II , Nitric Oxide Synthase Type III , Regeneration , Stem Cells/drug effects
14.
Circulation ; 105(17): 2012-8, 2002 Apr 30.
Article in English | MEDLINE | ID: mdl-11980678

ABSTRACT

BACKGROUND: This phase 1/2 study investigated the safety of percutaneous catheter-based gene transfer of naked plasmid DNA encoding for vascular endothelial growth factor 2 (phVEGF2) to left ventricular (LV) myocardium in a prospective, randomized, double-blind, placebo-controlled, dose-escalating study of inoperable patients with class III or IV angina. METHODS AND RESULTS: A steerable deflectable 8F catheter with a 27-gauge needle at its distal tip was advanced percutaneously to the endocardial surface of the LV in 19 patients (age, 61+/-2 years) with chronic myocardial ischemia who were not candidates for conventional revascularization. Patients were randomized in a double-blind fashion to receive 6 injections (total volume, 6.0 mL) of placebo or phVEGF2 in doses of 200 microg (n=9), 800 microg (n=9), or 2000 microg (n=1) guided by LV electromechanical (NOGA) mapping with a gene-to-placebo ratio of 2:1. A total of 114 LV injections were delivered and caused no hemodynamic alterations, sustained ventricular arrhythmias, ECG evidence of infarction, or ventricular perforation. End-point analysis at 12 weeks disclosed a statistically significant improvement in Canadian Cardiovascular Society (CCS) angina class in phVEGF2-treated versus placebo-treated patients (-1.3 versus -0.1, P=0.04). Remaining efficacy end points--including change in exercise duration (91.8 versus 3.9 seconds), functional improvement by > or =2 CCS classes (9 of 12 versus 1 of 6), and Seattle Angina Questionnaire data--all showed strong trends favoring efficacy of phVEGF2 versus placebo treatment. CONCLUSIONS: This phase 1/2, double-blind, randomized trial provides preliminary data that support safety of phVEGF2 catheter-mediated myocardial gene transfer. The statistically significant reduction in anginal class and strong positive trends for remaining end points suggest that a larger phase 2/3 trial is warranted.


Subject(s)
Endothelial Growth Factors/genetics , Genetic Therapy , Heart Ventricles , Lymphokines/genetics , Myocardial Ischemia/therapy , Angina Pectoris/diagnosis , Angina Pectoris/diagnostic imaging , Angina Pectoris/therapy , Body Surface Potential Mapping , Catheterization , Chronic Disease , Double-Blind Method , Exercise Test , Female , Genetic Therapy/adverse effects , Humans , Male , Middle Aged , Myocardial Ischemia/diagnosis , Myocardial Ischemia/diagnostic imaging , Tomography, Emission-Computed, Single-Photon , Treatment Outcome , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
15.
Circulation ; 105(3): 360-6, 2002 Jan 22.
Article in English | MEDLINE | ID: mdl-11804993

ABSTRACT

BACKGROUND: Kinins are modulators of cardiovascular function. After ischemic injury, enhanced kinin generation may contribute in processes responsible for tissue healing. METHODS AND RESULTS: Using pharmacological and genetic approaches, we investigated the role of kinin B(1) receptor in reparative angiogenesis in a murine model of limb ischemia. The effect of B(1) pharmacological manipulation on human endothelial cell proliferation and apoptosis was also studied in vitro. Abrogation of B(1) signaling dramatically inhibited the native angiogenic response to ischemia, severely compromising blood perfusion recovery. Outcome was especially impaired in B(1) knockouts that showed a very high incidence of limb necrosis, eventually leading to spontaneous auto-amputation. Conversely, local delivery of a long-acting B(1) receptor agonist enhanced collateral vascular growth in ischemic skeletal muscle, accelerated the rate of perfusion recovery, and improved limb salvage. In vitro, B(1) activation stimulated endothelial cell proliferation and survival, whereas B(1) antagonism induced apoptosis. CONCLUSIONS: Our results indicate that the B(1) plays an essential role in the host defense response to ischemic injury. B(1) signaling potentiation might be envisaged as a utilitarian target for the treatment of ischemic vascular disease.


Subject(s)
Bradykinin/analogs & derivatives , Ischemia/therapy , Neovascularization, Physiologic , Receptors, Bradykinin/agonists , Animals , Apoptosis , Bradykinin/pharmacology , Bradykinin Receptor Antagonists , Cell Division , Cells, Cultured , Endothelium, Vascular/cytology , Extremities/blood supply , Extremities/pathology , Humans , Ischemia/blood , Ischemia/pathology , Laser-Doppler Flowmetry , Mice , Mice, Knockout , Muscle, Skeletal/blood supply , Perfusion , Receptor, Bradykinin B1 , Receptors, Bradykinin/genetics , Signal Transduction
16.
Circulation ; 107(3): 461-8, 2003 Jan 28.
Article in English | MEDLINE | ID: mdl-12551872

ABSTRACT

BACKGROUND: We investigated whether catheter-based, intramyocardial transplantation of autologous endothelial progenitor cells can enhance neovascularization in myocardial ischemia. METHODS AND RESULTS: Myocardial ischemia was induced by placement of an ameroid constrictor around swine left circumflex artery. Four weeks after constrictor placement, CD31+ mononuclear cells (MNCs) were freshly isolated from the peripheral blood of each animal. After overnight incubation of CD31+ MNCs in noncoated plates, nonadhesive cells (NA/CD31+ MNCs) were harvested as the endothelial progenitor cell-enriched fraction. Nonadhesive CD31- cells (NA/CD31- MNCs) were also prepared. Autologous transplantation of 10(7) NA/CD31+ MNCs, 10(7) NA/CD31- MNCs, or PBS was performed with a NOGA mapping injection catheter to target ischemic myocardium. In a parallel study, 10(5) human CD34+ MNCs, 10(5) human CD34- MNCs, or PBS was transplanted into ischemic myocardium of nude rats 10 minutes after ligation of the left anterior descending coronary artery. In the swine study, ischemic area by NOGA mapping, Rentrop grade angiographic collateral development, and echocardiographic left ventricular ejection fraction improved significantly 4 weeks after transplantation of NA/CD31+ MNCs but not after injection of NA/CD31- MNCs or PBS. Capillary density in ischemic myocardium 4 weeks after transplantation was significantly greater in the NA/CD31+ MNC group than the control groups. In the rat study, echocardiographic left ventricular systolic function and capillary density were significantly better preserved in the CD34+ MNC group than in the control groups 4 weeks after myocardial ischemia. CONCLUSIONS: These favorable outcomes encourage future clinical trials of catheter-based, intramyocardial transplantation of autologous CD34+ MNCs in the setting of chronic myocardial ischemia.


Subject(s)
Endothelium, Vascular/cytology , Myocardial Ischemia/therapy , Myocardium/cytology , Neovascularization, Physiologic , Stem Cell Transplantation/methods , Animals , Antigens, CD34/analysis , Cardiac Catheterization , Cell Differentiation , Cell Lineage , Chronic Disease , Coronary Angiography , Fibrosis , Male , Myocardial Ischemia/diagnosis , Myocardial Ischemia/diagnostic imaging , Rats , Rats, Nude , Stem Cells/chemistry , Stem Cells/cytology , Stem Cells/physiology , Swine , Ultrasonography , Ventricular Function, Left
17.
Circulation ; 110(1): 36-45, 2004 Jul 06.
Article in English | MEDLINE | ID: mdl-15210598

ABSTRACT

BACKGROUND: Drug-eluting stents represent a useful strategy for the prevention of restenosis using various antiproliferative drugs. These strategies share the liability of impairing endothelial recovery, thereby altering the natural biology of the vessel wall and increasing the associated risk of stent thrombosis. Accordingly, we tested the hypothesis that local delivery via gene-eluting stent of naked plasmid DNA encoding for human vascular endothelial growth factor (VEGF)-2 could achieve similar reductions in neointima formation while accelerating, rather than inhibiting, reendothelialization. METHODS AND RESULTS: phVEGF 2-plasmid (100 or 200 microg per stent)-coated BiodivYsio phosphorylcholine polymer stents versus uncoated stents were deployed in a randomized, blinded fashion in iliac arteries of 40 normocholesterolemic and 16 hypercholesterolemic rabbits. Reendothelialization was nearly complete in the VEGF stent group after 10 days and was significantly greater than in control stents (98.7+/-1% versus 79.0+/-6%, P<0.01). At 3 months, intravascular ultrasound analysis revealed that lumen cross-sectional area (4.2+/-0.4 versus 2.27+/-0.3 mm(2), P<0.001) was significantly greater and percent cross-sectional narrowing was significantly lower (23.4+/-6 versus 51.2+/-10, P<0.001) in VEGF stents compared with control stents implanted in hypercholesterolemic rabbits. Transgene expression was detectable in the vessel wall along with improved functional recovery of stented segments, resulting in a 2.4-fold increase in NO production. CONCLUSIONS: Acceleration of reendothelialization via VEGF-2 gene-eluting stents provides an alternative treatment strategy for the prevention of restenosis. VEGF-2 gene-eluting stents may be considered as a stand-alone or combination therapy.


Subject(s)
Arterial Occlusive Diseases/prevention & control , Gene Transfer Techniques , Genetic Therapy/methods , Stents , Vascular Endothelial Growth Factors/genetics , Animals , Arterial Occlusive Diseases/diagnostic imaging , Arterial Occlusive Diseases/pathology , Combined Modality Therapy , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Gene Expression , Male , Nitric Oxide/biosynthesis , Plasmids/administration & dosage , Rabbits , Stem Cells/cytology , Ultrasonography , Vascular Endothelial Growth Factors/analysis
18.
Arterioscler Thromb Vasc Biol ; 22(6): 901-6, 2002 Jun 01.
Article in English | MEDLINE | ID: mdl-12067896

ABSTRACT

Vascular endothelial growth factor (VEGF) promotes angiogenesis by a variety of mechanisms including stimulation of endothelial cell proliferation and migration and increasing vascular permeability. Although its mitogenic activity is mediated primarily by the beta(2)-isoforms of protein kinase C (PKC), little is known about the signaling pathways transducing its other physiological properties. Accordingly, we used a novel inhibitor molecule to examine the role of PKC isoforms alpha and beta in mediating VEGF-induced angiogenesis and vascular permeability. Because conventional inhibitors of PKC, such as staurosporine or calphostin C, also inhibit a variety of other protein kinases, we used a novel compound to specifically inhibit PKC. A myristoylated peptide, which mimics the pseudosubstrate motif of PKC-alpha and -beta subtypes, has been shown to be a highly selective and cell-permeable inhibitor of PKC. Blocking led, as expected, to abrogation of VEGF-induced endothelial cell proliferation in vitro. In vivo, VEGF-induced angiogenesis was impaired by myristoylated peptide. Surprisingly, selective inhibition of PKC induced vascular permeability in vivo via a NO-dependent mechanism. Moreover, PKC inhibition led to a 6.4-fold induction of NO synthase (NOS) activity in endothelial cells. Our findings demonstrate that activation of PKC is a major signaling pathway required for VEGF-induced proliferation and angiogenesis, whereas vascular permeability was enhanced by blocking PKC. Inhibition of calcium-dependent PKC by itself led to induction of NOS. Although NOS is a downstream target for VEGF-induced angiogenesis, its induction by PKC inhibition was not sufficient to promote neovascularization. These results reveal that angiogenesis and vascular permeability induced by VEGF are mediated by mechanisms which ultimately diverge.


Subject(s)
Capillary Permeability/physiology , Endothelial Growth Factors/antagonists & inhibitors , Endothelial Growth Factors/physiology , Lymphokines/antagonists & inhibitors , Lymphokines/physiology , Neovascularization, Physiologic/physiology , Nitric Oxide/metabolism , Protein Kinase C/antagonists & inhibitors , Signal Transduction/physiology , Animals , Aorta/cytology , Brain/enzymology , Capillary Permeability/drug effects , Cattle , Cell Division/drug effects , Cells, Cultured , Cornea/blood supply , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/enzymology , Enzyme Inhibitors/pharmacology , Humans , Mice , Nitric Oxide Synthase/physiology , Rats , Umbilical Veins/cytology , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
19.
Hum Gene Ther ; 13(13): 1595-603, 2002 Sep 01.
Article in English | MEDLINE | ID: mdl-12228014

ABSTRACT

The purpose of this study was to document the kinetics of vascular endothelial growth factor (VEGF) protein release into the systemic circulation after phVEGF gene transfer for therapeutic angiogenesis. VEGF plasma levels were measured by ELISA in 64 patients undergoing gene transfer of plasmid DNA: intramuscular in 34 patients with peripheral artery disease, and intramyocardial in 30 patients with coronary disease. Baseline plasma VEGF was highly variable and not normally distributed. After intramuscular gene transfer, median plasma VEGF rose slightly, although significantly, by 7 days (38 to 41 pg/ml, p < 0.05), but was not different from baseline at 14, 21, or 28 days. After intramyocardial gene transfer, median plasma VEGF levels were significantly elevated compared with baseline on days 2, 3, and 7 (39, 38, and 45 pg/ml, respectively, each p < 0.05 vs. baseline value of 21 pg/ml). Day 7 plasma levels did not differ significantly as a function of phVEGF dose, or between intramyocardial and intramuscular injections (1.8 and 1.3 times baseline levels, respectively, p = 0.6), despite an almost 10-fold difference in mean phVEGF dose. Intramuscular and intramyocardial phVEGF injections result in significant, although modest, elevations of circulating gene product for <14 days, with no relationship to injected dose. While a statistically significant increase in circulating VEGF level can provide evidence of successful gene transfer for groups of patients, interpretation of results for individual subjects is complicated by wide variation in baseline VEGF and low circulating levels compared with baseline after gene transfer.


Subject(s)
Endothelial Growth Factors/blood , Endothelial Growth Factors/genetics , Gene Transfer Techniques , Genetic Vectors , Intercellular Signaling Peptides and Proteins/blood , Intercellular Signaling Peptides and Proteins/genetics , Lymphokines/blood , Lymphokines/genetics , Plasmids , Aged , Coronary Artery Disease/genetics , Coronary Artery Disease/therapy , Female , Genetic Therapy , Humans , Ischemia/therapy , Male , Middle Aged , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
20.
Neurobiol Aging ; 25(10): 1361-8, 2004.
Article in English | MEDLINE | ID: mdl-15465634

ABSTRACT

The physiologic ability of peripheral nerves to regenerate after injury is impaired with aging. However, the mechanisms responsible for this phenomenon are still incompletely characterized. In this study, we investigated whether aging influences the intraneural angiogenic response that occurs after injury and during regeneration of peripheral nerves. We performed a crush injury of the sciatic nerve in old and senescence accelerated mice and found that the peripheral nerves of these animals are unable to locally upregulate vascular endothelial growth factor (VEGF), a prototypical angiogenic cytokine, after injury and have substantial deficits in mounting an appropriate intraneural angiogenic response during nerve regeneration. Our findings provide new evidence of possible interdependent relationships between aging, VEGF, angiogenesis, and nerve regeneration and suggest that vascular abnormalities might play a role in aging-associated neurological dysfunction, with potentially important fundamental and clinical implications.


Subject(s)
Aging/metabolism , Neovascularization, Physiologic/physiology , Nerve Regeneration/physiology , Sciatic Nerve/injuries , Sciatic Nerve/physiopathology , Vascular Endothelial Growth Factor A/metabolism , Aging/pathology , Animals , Male , Mice , Mice, Inbred C57BL , Neural Conduction/physiology , Peripheral Nerve Injuries , Peripheral Nerves/blood supply , Peripheral Nerves/pathology , Peripheral Nerves/physiopathology , Recovery of Function/physiology , Sciatic Nerve/blood supply , Sciatic Nerve/pathology
SELECTION OF CITATIONS
SEARCH DETAIL