Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 28
1.
J Cereb Blood Flow Metab ; 40(2): 404-419, 2020 02.
Article En | MEDLINE | ID: mdl-30621518

The myoblast-mediated delivery of angiogenic genes represents a cell-based approach for targeted induction of therapeutic collateralization. Here, we tested the superiority of myoblast-mediated co-delivery of vascular endothelial growth factor-A (VEGF) together with platelet-derived growth factor-BB (PDGF-BB) on transpial collateralization of an indirect encephalomyosynangiosis (EMS) in a model of chronic cerebral ischemia. Mouse myoblasts expressing a reporter gene alone (empty vector), VEGF, PDGF-BB or VEGF and PDGF-BB through a single bi-cistronic vector (VIP) were implanted into the temporalis muscle of an EMS following permanent ipsilateral internal carotid artery occlusion in adult, male C57BL/6N mice. Over 84 days, myoblast engraftment and gene product expression, hemodynamic impairment, transpial collateralization, angiogenesis, pericyte recruitment and post-ischemic neuroprotection were assessed. By day 42, animals that received PDGF-BB in combination with VEGF (VIP) showed superior hemodynamic recovery, EMS collateralization and ischemic protection with improved pericyte recruitment around the parenchymal vessels and EMS collaterals. Also, supplementation of PDGF-BB resulted in a striking astrocytic activation with intrinsic VEGF mobilization in the cortex below the EMS. Our findings suggest that EMS surgery together with myoblast-mediated co-delivery of VEGF/PDGF-BB may have the potential to serve as a novel treatment strategy for augmentation of collateral flow in the chronically hypoperfused brain.


Becaplermin , Brain Ischemia , Cerebral Cortex , Cerebrovascular Circulation , Genetic Vectors , Myoblasts , Vascular Endothelial Growth Factor A , Animals , Becaplermin/biosynthesis , Becaplermin/genetics , Brain Ischemia/genetics , Brain Ischemia/metabolism , Brain Ischemia/physiopathology , Brain Ischemia/therapy , Cerebral Cortex/blood supply , Cerebral Cortex/metabolism , Chronic Disease , Male , Mice , Muscle, Skeletal/metabolism , Myoblasts/metabolism , Myoblasts/transplantation , Vascular Endothelial Growth Factor A/biosynthesis , Vascular Endothelial Growth Factor A/genetics
2.
Front Microbiol ; 8: 2130, 2017.
Article En | MEDLINE | ID: mdl-29163418

Influenza is a respiratory disease that causes annual epidemics. Antiviral treatment options targeting the virus exist, but their efficiency is limited and influenza virus strains easily develop resistance. Thus, new treatment strategies are urgently needed. In the present study, we investigated the anti-influenza virus properties of D,L-lysine acetylsalicylate ⋅ glycine (BAY 81-8781; LASAG) that is approved as Aspirin i.v. for intravenous application. Instead of targeting the virus directly BAY 81-8781 inhibits the activation of the NF-κB pathway, which is required for efficient influenza virus propagation. Using highly pathogenic avian influenza virus strains we could demonstrate that BAY 81-8781 was able to control influenza virus infection in vitro. In the mouse infection model, inhalation of BAY 81-8781 resulted in reduced lung virus titers and protection of mice from lethal infection. Pharmacological studies demonstrated that the oral route of administration was not suitable to reach the sufficient concentrations of BAY 81-8781 for a successful antiviral effect in the lung. BAY 81-8781 treatment of mice infected with influenza virus started as late as 48 h after infection was still effective in protecting 50% of the animals from death. In summary, the data represent a successful proof of the novel innovative antiviral concept of targeting a host cell signaling pathway that is required for viral propagation instead of viral structures.

3.
Stroke ; 46(1): 203-11, 2015 Jan.
Article En | MEDLINE | ID: mdl-25388423

BACKGROUND AND PURPOSE: Direct extracranial-intracranial bypass surgery for treatment of cerebral hemodynamic compromise remains hindered by complications but alternative simple and safe indirect revascularization procedures, such as an encephalomyosynangiosis (EMS), lack hemodynamic efficiency. Here, the myoblast-mediated transfer of angiogenic genes presents an approach for induction of therapeutic collateralization. In this study, we tested the effect of myoblast-mediated delivery of vascular endothelial growth factor-A (VEGF) to the muscle/brain interface of an EMS in a model of chronic cerebral hypoperfusion. METHODS: Permanent unilateral internal carotid artery-occlusion was performed in adult C57/BL6 mice with or without (no EMS) surgical grafting of an EMS followed by implantation of monoclonal mouse myoblasts expressing either VEGF164 or an empty vector (EV). Cerebral hemodynamic impairment, transpial collateralization, angiogenesis, mural cell investment, microvascular permeability, and cortical infarction after ipsilateral stroke were assessed by real-time laser speckle blood flow imaging, 2- and 3-dimensional immunofluorescence and MRI. RESULTS: VEGF-expressing myoblasts improved hemodynamic rescue by day 14 (no EMS 37±21%, EV 42±9%, VEGF 48±12%; P<0.05 for VEGF versus no EMS and versus EV), together with the EMS take rate (VEGF 60%, EV 18.2%; P<0.05) and angiogenesis of mature cortical microvessels below the EMS (P<0.05 for VEGF versus EV). Importantly, functional and morphological results were paralleled by a 25% reduction of cortical infarction after experimental stroke on the side of the EMS. CONCLUSIONS: Myoblast-mediated VEGF supplementation at the target site of an EMS could help overcome the clinical dilemma of poor surgical revascularization results and provide protection from ischemic stroke.


Brain Ischemia , Cerebral Revascularization , Cerebrovascular Circulation/genetics , Collateral Circulation/genetics , Genetic Therapy/methods , Myoblasts , Neovascularization, Physiologic/genetics , Vascular Endothelial Growth Factor A/genetics , Animals , Cerebrovascular Disorders , Chronic Disease , Genetic Vectors , Mice , Mice, Inbred C57BL
4.
PLoS One ; 8(6): e63504, 2013.
Article En | MEDLINE | ID: mdl-23785394

BACKGROUND: A potential role for coagulation factors in pulmonary arterial hypertension has been recently described, but the mechanism of action is currently not known. Here, we investigated the interactions between thrombin and the nitric oxide-cGMP pathway in pulmonary endothelial cells and experimental pulmonary hypertension. PRINCIPAL FINDINGS: Chronic treatment with the selective thrombin inhibitor melagatran (0.9 mg/kg daily via implanted minipumps) reduced right ventricular hypertrophy in the rat monocrotaline model of experimental pulmonary hypertension. In vitro, thrombin was found to have biphasic effects on key regulators of the nitric oxide-cGMP pathway in endothelial cells (HUVECs). Acute thrombin stimulation led to increased expression of the cGMP-elevating factors endothelial nitric oxide synthase (eNOS) and soluble guanylate cyclase (sGC) subunits, leading to increased cGMP levels. By contrast, prolonged exposition of pulmonary endothelial cells to thrombin revealed a characteristic pattern of differential expression of the key regulators of the nitric oxide-cGMP pathway, in which specifically the factors contributing to cGMP elevation (eNOS and sGC) were reduced and the cGMP-hydrolyzing PDE5 was elevated (qPCR and Western blot). In line with the differential expression of key regulators of the nitric oxide-cGMP pathway, a reduction of cGMP by prolonged thrombin stimulation was found. The effects of prolonged thrombin exposure were confirmed in endothelial cells of pulmonary origin (HPAECs and HPMECs). Similar effects could be induced by activation of protease-activated receptor-1 (PAR-1). CONCLUSION: These findings suggest a link between thrombin generation and cGMP depletion in lung endothelial cells through negative regulation of the nitric oxide-cGMP pathway, possibly mediated via PAR-1, which could be of relevance in pulmonary arterial hypertension.


Cyclic GMP/metabolism , Endothelial Cells/metabolism , Hypertension, Pulmonary/metabolism , Metabolic Networks and Pathways , Nitric Oxide/metabolism , Thrombin/metabolism , Animals , Cyclic Nucleotide Phosphodiesterases, Type 5/metabolism , Disease Models, Animal , Endothelial Cells/drug effects , Humans , Hypertension, Pulmonary/complications , Hypertension, Pulmonary/drug therapy , Hypertrophy, Right Ventricular/drug therapy , Hypertrophy, Right Ventricular/etiology , Male , Metabolic Networks and Pathways/drug effects , Nitric Oxide Synthase Type III/metabolism , Rats , Receptor, PAR-1/metabolism , Thrombin/administration & dosage , Thrombin/pharmacology
5.
FASEB J ; 26(6): 2486-97, 2012 Jun.
Article En | MEDLINE | ID: mdl-22391130

Therapeutic angiogenesis by delivery of vascular growth factors is an attractive strategy for treating debilitating occlusive vascular diseases, yet clinical trials have thus far failed to show efficacy. As a result, limb amputation remains a common outcome for muscle ischemia due to severe atherosclerotic disease, with an overall incidence of 100 per million people in the United States per year. A challenge has been that the angiogenic master regulator vascular endothelial growth factor (VEGF) induces dysfunctional vessels, if expressed outside of a narrow dosage window. We tested the hypothesis that codelivery of platelet-derived growth factor-BB (PDGF-BB), which recruits pericytes, could induce normal angiogenesis in skeletal muscle irrespective of VEGF levels. Coexpression of VEGF and PDGF-BB encoded by separate vectors in different cells or in the same cells only partially corrected aberrant angiogenesis. In marked contrast, coexpression of both factors in every cell at a fixed relative level via a single bicistronic vector led to robust, uniformly normal angiogenesis, even when VEGF expression was high and heterogeneous. Notably, in an ischemic hindlimb model, single-vector expression led to efficient growth of collateral arteries, revascularization, increased blood flow, and reduced tissue damage. Furthermore, these results were confirmed in a clinically applicable gene therapy approach by adenoviral-mediated delivery of the bicistronic vector. We conclude that coordinated expression of VEGF and PDGF-BB via a single vector constitutes a novel strategy for harnessing the potency of VEGF to induce safe and efficacious angiogenesis.


Neovascularization, Physiologic/physiology , Proto-Oncogene Proteins c-sis/therapeutic use , Vascular Endothelial Growth Factor A/therapeutic use , Adenoviridae/genetics , Animals , Becaplermin , Gene Transfer Techniques , Genetic Therapy/methods , Genetic Vectors , HEK293 Cells , Hindlimb/blood supply , Humans , Male , Mice , Mice, SCID , Muscle, Skeletal/blood supply , Platelet-Derived Growth Factor/therapeutic use , Proto-Oncogene Proteins c-sis/administration & dosage , Vascular Endothelial Growth Factor A/administration & dosage
6.
Ann Rheum Dis ; 71(6): 1019-26, 2012 Jun.
Article En | MEDLINE | ID: mdl-22294631

BACKGROUND: Fibrosis and vascular disease are cardinal features of systemic sclerosis (SSc). Stimulators of soluble guanylate cyclase (sGC) are vasoactive drugs that are currently being evaluated in phase III clinical trials for pulmonary arterial hypertension. OBJECTIVE: To study the antifibrotic potency of sGC stimulators. METHODS: The effect of the sGC stimulator BAY 41-2272 on the release of collagen from dermal fibroblasts was examined. The antifibrotic effects of BAY 41-2272 on prevention and regression of fibrosis in bleomycin-induced dermal fibrosis and in Tsk-1 mice were also studied. Telemetric blood pressure studies in conscious mice were used to study potential hypotensive effects of sGC stimulation. RESULTS: sGC stimulation with BAY 41-2272 dose-dependently inhibited collagen release in dermal fibroblasts from patients with SSc and healthy individuals. Furthermore, BAY 41-2272 stopped the development of bleomycin-induced dermal fibrosis and skin fibrosis in Tsk-1 mice, preventing dermal and hypodermal thickening, reducing the numbers of myofibroblasts and reducing the hydroxyproline content. In addition, BAY 41-2272 was highly effective in the treatment of established fibrosis in the modified models of bleomycin-induced skin fibrosis and Tsk-1 mice. Treatment with sGC stimulators was well tolerated. Relevant antifibrotic doses of BAY 41-2272 did not affect systemic blood pressure and heart rate in mice. CONCLUSIONS: These findings demonstrate potent antifibrotic effects and good tolerability of sGC stimulators in various experimental models of SSc. Given their potential vasoactive properties, sGC stimulators may be promising candidates for the dual treatment of fibrosis and vascular disease in SSc.


Dermis/cytology , Fibroblasts/drug effects , Pyrazoles/pharmacology , Pyridines/pharmacology , Receptors, Cytoplasmic and Nuclear/agonists , Scleroderma, Systemic/drug therapy , Animals , Antibiotics, Antineoplastic/pharmacology , Bleomycin/pharmacology , Blood Pressure/drug effects , Cells, Cultured , Collagen/metabolism , Disease Models, Animal , Enzyme Activation/drug effects , Fibroblasts/enzymology , Fibroblasts/pathology , Fibrosis/drug therapy , Fibrosis/metabolism , Fibrosis/pathology , Guanylate Cyclase/metabolism , Humans , Mice , Mice, Mutant Strains , Protein Serine-Threonine Kinases/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Scleroderma, Systemic/metabolism , Scleroderma, Systemic/pathology , Soluble Guanylyl Cyclase
7.
J Neurosci Methods ; 201(1): 61-6, 2011 Sep 30.
Article En | MEDLINE | ID: mdl-21835200

An encephalomyosynangiosis (EMS) is a temporal muscle graft that is placed onto the surface of the brain to serve as a source for collateral vessel growth for brain revascularization in patients with Moyamoya Disease (MMD). To facilitate an EMS in patients with occlusive cerebrovascular diseases other than MMD, the transfer of pro-angiogenic genes via transplantation of retrovirally transduced myoblasts into the temporal muscle may represent an innovative approach to augment collateralization. Thus, we tested whether retrovirally transfected myoblasts can spontaneously fuse with the non-ischemic and uninjured muscle tissue and if a reporter gene can be stably expressed within the temporal muscle of the EMS. Primary mouse myoblasts expressing a reporter gene were implanted into the temporal muscle prior to an EMS being performed on C57/BL6 mice. Three different implantation modalities were evaluated: (a) intramuscular injection, (b) application of a cell pellet and (c) a combination of both techniques. Myoblast implantation resulted in spontaneous fusion with the host muscle fibers and stable reporter gene expression at both the muscle/brain interface and within the non-ischemic and uninjured temporal muscle in all animals. The mean number of fused hybrid myofibers was 59±28 after injection, 37±30 after pellet application and 60±23 after a combination of both techniques. Regardless of the implantation modality, an abundant extracellular expression of the reporter gene was evident at the muscle/brain interface; in the case of myoblast delivery by injection, expression was also observed around the needle tract marking the implantation site. This method could be used in the future to deliver angiogenic growth factors to the muscle/brain interface in order to improve revascularization after an EMS.


Brain/physiology , Cerebral Revascularization/methods , Gene Transfer Techniques/trends , Genetic Therapy/trends , Myoblasts/physiology , Myoblasts/transplantation , Animals , Brain/blood supply , Brain/surgery , Cell Transplantation/trends , Male , Mice , Mice, Inbred C57BL , Moyamoya Disease/genetics , Moyamoya Disease/therapy , Temporal Muscle/physiology , Temporal Muscle/transplantation
8.
Cardiovasc Res ; 92(1): 159-68, 2011 Oct 01.
Article En | MEDLINE | ID: mdl-21676958

AIMS: Anticoagulation with warfarin is recommended for the treatment of patients with pulmonary arterial hypertension (PAH). However, the therapeutic benefit of anticoagulation has not yet been demonstrated experimentally or clinically. Here, rivaroxaban, an oral, direct factor Xa (FXa) inhibitor, was compared with warfarin and enoxaparin in the prevention of right ventricular (RV) dysfunction and hypertrophy in the monocrotaline (MCT) model of pulmonary hypertension. METHODS AND RESULTS: Sprague-Dawley rats (n = 10 per group) were randomized to receive rivaroxaban, warfarin, enoxaparin, or placebo before receiving a subcutaneous injection of MCT 60 mg/kg or saline. Rivaroxaban and enoxaparin were administered for 28 days starting 4 h before MCT injection; warfarin was given for 35 days initiated 7 days before MCT injection. RV haemodynamics and hypertrophy were assessed 28 days after MCT administration. Rivaroxaban dose-dependently reduced systolic and end-diastolic RV pressure increase and RV hypertrophy. Warfarin reduced RV pressure increase only. Enoxaparin had no effect on either parameter. Severe bleeding occurred in four and five rats treated with warfarin and enoxaparin, respectively, whereas no overt bleeding was observed in rats treated with rivaroxaban. CONCLUSION: Selective, direct inhibition of FXa by rivaroxaban effectively prevented RV dysfunction and hypertrophy in MCT-injected rats, indicating a role for coagulation factors in experimental pulmonary hypertension. Clinical investigation of the impact of early and continued administration of a specific FXa inhibitor such as rivaroxaban on the course of PAH should be considered.


Factor Xa/physiology , Hypertension, Pulmonary/etiology , Animals , Blood Coagulation , Enoxaparin/pharmacology , Factor Xa Inhibitors , Familial Primary Pulmonary Hypertension , Hemodynamics/drug effects , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/drug therapy , Hypertrophy, Right Ventricular/prevention & control , Male , Monocrotaline , Morpholines/pharmacology , Rats , Rats, Sprague-Dawley , Rats, Wistar , Rivaroxaban , Thiophenes/pharmacology , Thrombosis/etiology , Warfarin/pharmacology
9.
Thromb Haemost ; 102(5): 892-9, 2009 Nov.
Article En | MEDLINE | ID: mdl-19888525

Heparins, either unfractionated or low-molecular-weight (UFH and LMWHs), and vitamin K antagonists (VKAs) are currently the anticoagulants of choice for the prevention of post-operative venous thromboembolism (VTE) and for the treatment of acute venous and arterial thromboembolism. While VKAs are widely used in the US, LMWHs are the standard of care in the EU. Although efficacious, these agents are associated with a number of drawbacks, such as the risk of heparin-induced thrombocytopenia, the need for frequent coagulation monitoring in the case of UFH and VKAs, and the parenteral mode of administration in the case of heparins, which can lead to problems associated with patient compliance. There is a need for new anticoagulants that overcome these limitations. Direct, small-molecule inhibitors of coagulation proteins targeting a single enzyme in the coagulation cascade - particularly thrombin or Factor Xa - have been developed in recent years. Two agents, the direct thrombin inhibitor dabigatran and the direct Factor Xa inhibitor rivaroxaban, have recently been approved in the EU and several other countries for the prevention of VTE after total hip or knee replacement surgery. Here we will review data that suggest that the antithrombin-independent mechanism of action of these agents, particularly that of direct Factor Xa inhibitors, leads to increased efficacy with similar safety profiles compared with the antithrombin-dependent heparins. Although the end of the heparins era is not to be expected, the new anticoagulants presented in this review potentially represent the future of anticoagulation.


Anticoagulants/pharmacology , Blood Coagulation Factors/antagonists & inhibitors , Thrombophilia/drug therapy , Anticoagulants/chemistry , Anticoagulants/therapeutic use , Antithrombin III/antagonists & inhibitors , Clinical Trials as Topic , Drug Design , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Factor V/antagonists & inhibitors , Factor Xa , Factor Xa Inhibitors , Hemorrhage/chemically induced , Heparin/pharmacology , Heparin/therapeutic use , Heparin, Low-Molecular-Weight/pharmacology , Heparin, Low-Molecular-Weight/therapeutic use , Humans , Molecular Weight , Postoperative Complications/chemically induced , Postoperative Complications/prevention & control , Purpura, Thrombocytopenic, Idiopathic/chemically induced , Purpura, Thrombocytopenic, Idiopathic/prevention & control , Venous Thromboembolism/prevention & control , Vitamin K/antagonists & inhibitors
10.
ALTEX ; 26(3): 177-85, 2009.
Article En | MEDLINE | ID: mdl-19907905

In vivo luminescent imaging technology was introduced in experimental life science research several years ago and has rapidly gained wide acceptance. By making use of this technology substantially more information can be gained from animal experiments than was previously possible. The concept of the 3Rs describes the aim to Refine, Reduce and Replace animal models in research. The goal of the present paper is to systematically investigate the impact of luminescent imaging on the 3Rs. In particular, three examples of applications are explained in detail so as to be accessible to the reader unfamiliar with the procedure. The examples are subsequently analysed for and categorised according to their concrete effect on animal welfare as defined by the 3Rs.


Animal Testing Alternatives/methods , Luminescent Measurements/methods , Animal Experimentation/ethics , Animals , Biomedical Research/ethics , Biomedical Research/methods , Cell- and Tissue-Based Therapy , Heart , Listeriosis/microbiology , Luminescent Measurements/instrumentation , Mice , Neoplasms
11.
Methods Mol Biol ; 574: 249-59, 2009.
Article En | MEDLINE | ID: mdl-19685314

Bioluminescence using the reporter enzyme firefly luciferase (Fluc) and the substrate luciferin enables non-invasive optical imaging of living animals with extremely high sensitivity. This type of analysis enables studies of gene expression, tumor growth, and cell migration over time in live animals that were previously not possible. However, a major limitation of this system is that Fluc activity is restricted to the intracellular environment, which precludes important applications of in vivo imaging such as antibody labeling, or serum protein monitoring. In order to expand the application of bioluminescence imaging to other enzymes, we characterized a sequential reporter-enzyme luminescence (SRL) technology for the in vivo detection of beta-galactosidase (beta-gal) activity. The substrate is a "caged" D-luciferin conjugate that must first be cleaved by beta-gal before it can be catalyzed by Fluc in the final, light-emitting step. Hence, luminescence is dependent on and correlates with beta-gal activity. A variety of experiments were performed in order to validate the system and explore potential new applications. We were able to visualize non-invasively over time constitutive beta-gal activity in engineered cells, as well as inducible tissue-specific beta-gal expression in transgenic mice. Since beta-gal, unlike Fluc, retains full activity outside of cells, we were able to show that antibodies conjugated to the recombinant beta-gal enzyme could be used to detect and localize endogenous cells and extracellular antigens in vivo. In addition, we developed a low-affinity beta-gal complementation system that enables inducible, reversible protein interactions to be monitored in real time in vivo, for example, sequential responses to agonists and antagonists of G-protein-coupled receptors (GPCRs). Thus, using SRL, the exquisite luminescent properties of Fluc can be combined with the advantages of another enzyme. Other substrates have been described that extend the scope to endogenous enzymes, such as cytochromes or caspases, potentially enabling additional unprecedented applications.


Genes, Reporter , Luminescence , beta-Galactosidase/metabolism , Animals , Mice , Mice, Nude , beta-Galactosidase/genetics
12.
Arterioscler Thromb Vasc Biol ; 28(9): 1614-20, 2008 Sep.
Article En | MEDLINE | ID: mdl-18511699

OBJECTIVE: The endogenous role of the VEGF family member vascular endothelial growth factor-B (VEGF-B) in pathological angiogenesis remains unclear. METHODS AND RESULTS: We studied the role of VEGF-B in various models of pathological angiogenesis using mice lacking VEGF-B (VEGF-B(-/-)) or overexpressing VEGF-B(167). After occlusion of the left coronary artery, VEGF-B deficiency impaired vessel growth in the ischemic myocardium whereas, in wild-type mice, VEGF-B(167) overexpression enhanced revascularization of the infarct and ischemic border zone. By contrast, VEGF-B deficiency did not affect vessel growth in the wounded skin, hypoxic lung, ischemic retina, or ischemic limb. Moreover, VEGF-B(167) overexpression failed to enhance vascular growth in the skin or ischemic limb. CONCLUSIONS: VEGF-B appears to have a relatively restricted angiogenic activity in the ischemic heart. These insights might offer novel therapeutic opportunities.


Coronary Vessels/metabolism , Ischemia/metabolism , Myocardial Ischemia/metabolism , Myocardium/metabolism , Neovascularization, Physiologic , Vascular Endothelial Growth Factor B/metabolism , Angiogenesis Inducing Agents/metabolism , Animals , Coronary Vessels/drug effects , Coronary Vessels/physiopathology , Disease Models, Animal , Gene Transfer Techniques , Genetic Therapy/methods , Hindlimb , Ischemia/pathology , Ischemia/physiopathology , Ischemia/therapy , Lung/blood supply , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , Muscle, Skeletal/blood supply , Myocardial Ischemia/pathology , Myocardial Ischemia/physiopathology , Myocardial Ischemia/therapy , Myocardium/pathology , Neovascularization, Physiologic/drug effects , Recombinant Proteins/metabolism , Retinal Vessels/metabolism , Skin/blood supply , Up-Regulation , Vascular Endothelial Growth Factor B/administration & dosage , Vascular Endothelial Growth Factor B/deficiency , Vascular Endothelial Growth Factor B/genetics , Vascular Endothelial Growth Factor Receptor-1/metabolism
13.
FASEB J ; 21(14): 3819-26, 2007 Dec.
Article En | MEDLINE | ID: mdl-17942828

G-protein coupled receptors (GPCRs) are a versatile and ubiquitous family of membrane receptors that transmit extracellular signals to mammalian cells and constitute the most important class of drug targets. Yet, sensitive and specific methods are lacking that would allow quantitative comparisons of pharmacologic properties of these receptors in physiological or pathological settings in live animals. We sought to overcome these limitations by employing low affinity, reversible beta-galactosidase complementation to quantify GPCR activation via interaction with beta-arrestin. A panel of cell lines was engineered expressing different GPCRs together with the reporter system. In vitro evaluation revealed highly sensitive, dynamic, and specific assessment of GPCR agonists and antagonists. Following implantation of the cells into mice, it was possible for the first time to monitor pharmacological GPCR activation and inhibition in their physiological context by noninvasive bioluminescence imaging in living animals. This technology has unique advantages that enable novel applications in the functional investigation of GPCR modulation in live animals in biological research and drug discovery.


Receptors, G-Protein-Coupled/metabolism , Animals , CHO Cells , COS Cells , Chlorocebus aethiops , Cricetinae , Cricetulus , Firefly Luciferin , Humans , Luminescent Agents , Mice , Mice, Inbred BALB C , Mice, Nude , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/genetics
14.
Nat Chem Biol ; 3(10): 668-77, 2007 Oct.
Article En | MEDLINE | ID: mdl-17828252

We have generated a series of quenched near-infrared fluorescent activity-based probes (qNIRF-ABPs) that covalently target the papain-family cysteine proteases shown previously to be important in multiple stages of tumorigenesis. These 'smart' probes emit a fluorescent signal only after covalently modifying a specific protease target. After intravenous injection of NIRF-ABPs into mice bearing grafted tumors, noninvasive, whole-body imaging allowed direct monitoring of cathepsin activity. Importantly, the permanent nature of the probes also allowed secondary, ex vivo biochemical profiling to identify specific proteases and to correlate their activity with whole-body images. Finally, we demonstrate that these probes can be used to monitor small-molecule inhibition of protease targets both biochemically and by direct imaging methods. Thus, NIRF-ABPs are (i) potentially valuable new imaging agents for disease diagnosis and (ii) powerful tools for preclinical and clinical testing of small-molecule therapeutic agents in vivo.


Cathepsins/analysis , Cysteine Endopeptidases/metabolism , Diagnostic Imaging/methods , Fluorescent Dyes , Whole Body Imaging/methods , Animals , Cathepsins/metabolism , Electrophoresis, Polyacrylamide Gel , Fluorescent Dyes/metabolism , Mice , Mice, Inbred BALB C , Microscopy, Fluorescence , Neoplasms/pathology , Neoplasms/ultrastructure , Sensitivity and Specificity , Time Factors
15.
J Heart Lung Transplant ; 26(3): 273-80, 2007 Mar.
Article En | MEDLINE | ID: mdl-17346630

BACKGROUND: Histology-based survival assessment of cell grafts does not allow for in vivo follow-up. In this study we introduce two new experimental models for longitudinal in vivo survival studies of cardiac cell grafts using optical bioluminescence imaging. METHODS: H9c2 cardiomyoblasts expressing both firefly luciferase (fluc) and green fluorescent protein (GFP) reporter genes were implanted into Lewis rats. In Model 1, H9c2-fluc-IRES-GFP cells (0.5 x 10(6)) were implanted into a cryoinjured abdominal wall muscle. Cells were injected using either liquid collagen (Matrigel [MG]) or phosphate-buffered saline (PBS) suspension. Cell survival was evaluated in vivo using bioluminescence imaging on days 1, 5 and 10 post-operatively. In model 2, rats underwent ligation of the left anterior descending (LAD) artery. The donor hearts were harvested, and the infarcted region was restored ex situ using 1 x 10(6) H9c2-fluc-IRES-GFP cells seeded in collagen matrix (Gelfoam [GF]) or suspended in PBS (n = 8/group). Hearts were then transplanted into the abdomen of syngeneic recipients. Optical bioluminescence imaging was performed on Days 1, 5, 8 and 14 post-operatively. After 4 weeks, immunohistologic studies were performed. RESULTS: For model 1, at day 5, bioluminescence signals were markedly higher for the H9c2/MG group (449 +/- 129 photons/second x 10(3)) compared with the H9c2/PBS group (137 +/- 82 photons/second x 10(3)) (p < 0.05). For model 2, bioluminescence signals were significantly (p < 0.04) higher in the H9c2/GF group compared with plain cell injection on days 5 (534 +/- 115 vs 219 +/- 34) and 8 (274 +/- 34 vs 180 +/- 23). Data were in accordance with GFP immunohistology. CONCLUSIONS: Optical bioluminescence is a powerful method for assessment of cardiac cell graft survival in vivo. Collagen matrices support early survival of cardiomyoblasts after transplantation into injured musculature.


Abdominal Muscles/surgery , Collagen/administration & dosage , Diagnostic Imaging , Laminin/administration & dosage , Luminescent Agents , Myoblasts, Cardiac/transplantation , Myocardial Infarction/surgery , Optics and Photonics , Proteoglycans/administration & dosage , Animals , Cell Line , Cell Survival/drug effects , Collagen/pharmacology , Drug Combinations , Green Fluorescent Proteins , Heart Transplantation , Injections , Laminin/pharmacology , Luciferases, Firefly , Male , Proteoglycans/pharmacology , Rats , Rats, Inbred Lew , Time Factors , Transplantation, Heterotopic , Transplantation, Isogeneic
16.
FASEB J ; 21(9): 2074-85, 2007 Jul.
Article En | MEDLINE | ID: mdl-17325231

The major vascular endothelial growth factor (VEGF) isoforms are splice variants from a single gene that differ in their extent of heparin affinity due to the absence of the heparin binding domain in the smallest isoform (mouse VEGF120, human VEGF121). A long-held assumption that has guided the use of VEGF isoforms clinically has been that their differences in heparin binding dictate their ability to diffuse through tissue, with VEGF121 moving most freely and that the distribution of recombinant VEGF would have therapeutically relevant consequences. To test this assumption, we delivered the genes encoding these isoforms by myoblast-mediated gene transfer, a means of delivering genes to highly localized sites within muscle. Surprisingly, all isoforms induced comparable extremely localized physiological effects. Significantly, irrespective of the isoform delivered, the vessels passing within several micrometers of muscle fibers expressing VEGF displayed sharply delineated changes in morphology. The induction of capillary wrapping around VEGF-producing fibers, and of vascular malformations in the muscle at high levels, did not differ among isoforms. These results indicate that heparin binding is not essential for the localization of VEGF in adult tissue and suggest that the preferential delivery of VEGF121 cDNA for clinical applications may not have a physiological basis.


Heparin/metabolism , Neovascularization, Physiologic/drug effects , Vascular Endothelial Growth Factor A/pharmacology , Alternative Splicing , Animals , Arterioles/drug effects , Arterioles/growth & development , Biological Availability , Capillaries/drug effects , Capillaries/growth & development , Cells, Cultured/metabolism , Cells, Cultured/transplantation , Cells, Cultured/virology , Diffusion , Ear, External/blood supply , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Hemangioma/etiology , Hemangioma/pathology , Male , Mice , Mice, SCID , Molecular Weight , Muscle Neoplasms/etiology , Muscle Neoplasms/pathology , Muscle, Skeletal/blood supply , Myoblasts/metabolism , Myoblasts/transplantation , Myoblasts/virology , Protein Binding , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Isoforms/pharmacology , RNA, Messenger/biosynthesis , Retroviridae/genetics , Transduction, Genetic , Transgenes , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
17.
FASEB J ; 20(14): 2657-9, 2006 Dec.
Article En | MEDLINE | ID: mdl-17095533

The critical role of vascular endothelial growth factor (VEGF) expression levels in developmental angiogenesis is well established. Nonetheless, the effects of different local (microenvironmental) VEGF concentrations in ischemia have not been studied in the adult organism, and VEGF delivery to patients has been disappointing. Here, we demonstrate the existence of both lower and upper threshold levels of microenvironmental VEGF concentrations for the induction of therapeutic vessel growth in ischemia. In the ischemic hind limb, implantation of myoblasts transduced to express VEGF164 at different levels per cell increased blood flow only moderately, and vascular leakage and aberrant preangiomatous vessels were always induced. When the same total dose was uniformly distributed by implanting a monoclonal population derived from a single VEGF-expressing myoblast, blood flow was fully restored to nonischemic levels, collateral growth was induced, and ischemic damage was prevented. Hemangiomas were avoided and only normal, pericyte-covered vessels were induced persisting over 15 mo. Surprisingly, clones uniformly expressing either lower or higher VEGF levels failed to provide any functional benefit. A biphasic effect of VEGF dose on vessel number and diameter was found. Blood flow was only improved if vessels were increased both in size and in number. Microenvironmental VEGF concentrations determine efficacy and safety in a therapeutic setting.


Ischemia/metabolism , Neovascularization, Physiologic/physiology , Vascular Endothelial Growth Factor A/metabolism , Animals , Gene Expression Regulation , Hindlimb/blood supply , Hindlimb/metabolism , Male , Mice , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Vascular Endothelial Growth Factor A/genetics
18.
Circulation ; 114(1 Suppl): I167-73, 2006 Jul 04.
Article En | MEDLINE | ID: mdl-16820568

BACKGROUND: Cardiac cell transplantation is limited by poor graft viability. We aimed to enhance the survival of transplanted cardiomyoblasts using growth factor-supplemented collagen matrices. METHODS AND RESULTS: H9c2 cardiomyoblasts were lentivirally transduced to express firefly luciferase and green fluorescent protein (GFP). Lewis rats underwent ligation of the left anterior descending artery (LAD) ligation to induce an anterior wall myocardial infarction. Hearts (n=9/group) were harvested and restored ex vivo with 1 x 10(6) genetically labeled H9c2 cells either in (1) saline-suspension, or seeded onto (2) collagen-matrix (Gelfoam [GF];), (3) GF/Matrigel (GF/MG), (4) GF/MG/VEGF (10 microg/mL), or (5) GF/MG/FGF (10 microg/mL). Hearts were then abdominally transplanted into syngeneic recipients (working heart model). Controls (n=6/group) underwent infarction followed by GF implantation or saline injection. Cell survival was evaluated using optical bioluminescence on days 1, 5, 8, 14, and 28 postoperatively. At 4 weeks, fractional shortening and ejection fraction were determined using echocardiography and magnetic resonance imaging, respectively. Graft characteristics were assessed by immunohistology. Bioluminescence signals on days 5, 8, and 14 were higher for GF-based grafts compared with plain H9c2 injections (P<0.03). Signals were higher for GF/MG grafts compared with GF alone (P<0.02). GFP-positive, spindle-shaped H9c2 cells were found integrated in the infarct border zones at day 28. Left ventricular (LV) function of hearts implanted with collagen-based grafts was better compared with controls (P<0.05). Vascular endothelial growth factor or fibroblast growth factor did not further improve graft survival or heart function. CONCLUSIONS: Collagen matrices enhance early survival of H9c2 cardiomyoblasts after transplantation into ischemic hearts and lead to improved LV function. Further optimization of the graft design should make restoration of large myocardial infarctions by tissue engineering approaches effective.


Collagen/pharmacology , Extracellular Matrix/transplantation , Graft Survival/drug effects , Heart Transplantation , Implants, Experimental , Myoblasts/transplantation , Myocardial Infarction/surgery , Myocytes, Cardiac/transplantation , Tissue Engineering , Abdominal Cavity , Animals , Cell Survival/drug effects , Drug Combinations , Fibroblast Growth Factors/pharmacology , Gelatin Sponge, Absorbable , Genes, Reporter , Laminin , Magnetic Resonance Imaging , Male , Myocardial Contraction , Myocardial Infarction/diagnostic imaging , Myocardial Infarction/pathology , Proteoglycans , Rats , Rats, Inbred Lew , Stroke Volume , Tissue Engineering/methods , Transduction, Genetic , Transplantation, Heterotopic , Transplantation, Isogeneic , Ultrasonography , Vascular Endothelial Growth Factor A/pharmacology , Ventricular Function, Left
19.
Circulation ; 114(1 Suppl): I174-80, 2006 Jul 04.
Article En | MEDLINE | ID: mdl-16820569

BACKGROUND: Cell transplantation for myocardial repair is limited by early cell death. Gene therapy with human Bcl-2 (hBcl-2) has been shown to attenuate apoptosis in the experimental setting. Therefore, we studied the potential benefit of hBcl-2 transgene expression on the survival of cardiomyoblast grafts in ischemic rat hearts. METHODS AND RESULTS: H9c2 rat cardiomyoblasts were genetically modified to express both firefly luciferase and green fluorescent protein (mH9c2). The cells were then transduced with adenovirus carrying hBcl-2 (AdCMVhBcl-2/mH9c2). Lewis rats underwent ligation of the left anterior descending artery (LAD) to induce a sizable left ventricular (LV) infarct. Hearts were explanted and the infarcted region was restored using collagen matrix (CM) seeded with 1x10(6) mH9c2 cells (n=9) or AdCMVhBcl-2/mH9c2 cells (n=9). Control animals received CM alone (n=6) or no infarct (n=6). Restored hearts were transplanted into the abdomen of syngeneic recipients in a "working heart" model. Cell survival was evaluated using optical bioluminescence imaging on days 1, 5, 8, 14, and 28 after surgery. The left heart function was assessed 4 weeks postoperatively using echocardiography and magnetic resonance imaging. During 4 weeks after surgery, the optical imaging signal for the AdCMVhBCL2/mH9c2 group was significantly (P<0.05) higher than that of the mH9c2-control group. Both grafts led to better fractional shortening (AdCMVhBcl-2/mH9c2: 0.21+/-0.03; mH9c2: 0.21+/-0.04; control: 0.15+/-0.03; P=0.04) and ejection fraction (AdCMVhBcl-2/mH9c2: 47.0+/-6.2; mH9c2: 48.7+/-6.1; control: 34.3+/-6.0; P=0.02) compared with controls. Importantly, no malignant cells were found in postmortem histology. CONCLUSIONS: Transduction of mH9c2 cardiomyoblasts with AdCMVhBcl-2 increased graft survival in ischemic rat myocardium without causing malignancies. Both AdCMVhBcl-2/mH9c2 and mH9c2 grafts improved LV function.


Genes, bcl-2 , Genetic Therapy , Myoblasts/transplantation , Myocardial Infarction/therapy , Myocytes, Cardiac/transplantation , Transgenes , Abdomen , Abdominal Injuries/pathology , Abdominal Injuries/therapy , Abdominal Wall/pathology , Adenoviridae/genetics , Animals , Apoptosis , Cold Temperature/adverse effects , Collagen/pharmacology , Defective Viruses/genetics , Genes, Reporter , Genetic Vectors/therapeutic use , Heart Transplantation , Humans , Male , Rats , Rats, Inbred Lew , Transplantation, Heterotopic , Ventricular Function, Left
20.
Nat Methods ; 3(4): 295-301, 2006 Apr.
Article En | MEDLINE | ID: mdl-16554835

We generated a sequential reporter-enzyme luminescence (SRL) technology for in vivo detection of beta-galactosidase (beta-gal) activity. The substrate, a caged D-luciferin-galactoside conjugate, must first be cleaved by beta-gal before it can be catalyzed by firefly luciferase (FLuc) to generate light. As a result, luminescence is dependent on beta-gal activity. Using this technology, constitutive beta-gal activity in engineered cells and inducible tissue-specific beta-gal expression in transgenic mice can now be visualized noninvasively over time. A substantial advantage of beta-gal as a bioluminescent probe is that the enzyme retains full activity outside of cells, unlike FLuc, which requires intracellular cofactors. As a result, antibodies conjugated to the recombinant beta-gal enzyme can be used to detect endogenous cells and extracellular antigens in vivo. Thus, coupling the properties of FLuc to the advantages of beta-gal permits bioluminescent imaging applications that previously were not possible.


Luminescent Measurements/methods , beta-Galactosidase/analysis , Animals , Catalysis , Cell Membrane Permeability , Gene Expression Regulation , Genes, Reporter , Light , Luciferases/genetics , Luciferases/metabolism , Lymphoid Tissue/cytology , Lymphoid Tissue/metabolism , Lymphoid Tissue/ultrastructure , Mice , Mice, Transgenic , Sensitivity and Specificity , beta-Galactosidase/metabolism
...