Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Eur Heart J ; 45(4): 255-264, 2024 Jan 27.
Article in English | MEDLINE | ID: mdl-37634134

ABSTRACT

BACKGROUND AND AIMS: Clinical management of critical limb-threatening ischaemia (CLTI) is focused on prevention and treatment of atherosclerotic arterial occlusions. The role of microvascular pathology in disease progression is still largely unspecified and more importantly not utilized for treatment. The aim of this explorative study was to characterize the role of the microvasculature in CLTI pathology. METHODS: Clinical high-resolution imaging of CLTI patients (n = 50) and muscle samples from amputated CLTI limbs (n = 40) were used to describe microvascular pathology of CLTI at the level of resting muscle blood flow and microvascular structure, respectively. Furthermore, a chronic, low arterial driving pressure-simulating ischaemia model in rabbits (n = 24) was used together with adenoviral vascular endothelial growth factor A gene transfers to study the effect of microvascular alterations on muscle outcome. RESULTS: Resting microvascular blood flow was not depleted but displayed decreased capillary transit time (P < .01) in CLTI muscles. Critical limb-threatening ischaemia muscle microvasculature also exhibited capillary enlargement (P < .001) and further arterialization along worsening of myofibre atrophy and detaching of capillaries from myofibres. Furthermore, CLTI-like capillary transformation was shown to worsen calf muscle force production (P < .05) and tissue outcome (P < .01) under chronic ischaemia in rabbits and in healthy, normal rabbit muscle. CONCLUSIONS: These findings depict a progressive, hypoxia-driven transformation of the microvasculature in CLTI muscles, which pathologically alters blood flow dynamics and aggravates tissue damage under low arterial driving pressure. Hypoxia-driven capillary enlargement can be highly important for CLTI outcomes and should therefore be considered in further development of diagnostics and treatment of CLTI.


Subject(s)
Peripheral Arterial Disease , Humans , Rabbits , Animals , Peripheral Arterial Disease/therapy , Risk Factors , Vascular Endothelial Growth Factor A , Ischemia , Hypoxia , Treatment Outcome , Retrospective Studies , Chronic Disease
2.
Cells ; 12(16)2023 08 14.
Article in English | MEDLINE | ID: mdl-37626870

ABSTRACT

This study aimed to show the significance of capillary function in post-ischemic recovery from the perspective of physiological parameters, such as blood flow, hemoglobin oxygenation and tissue regeneration. Muscle-level microvascular alterations of blood flow and hemoglobin oxygenation, and post-ischemic myofiber and capillary responses were analyzed in aged, healthy C57Bl/6J mice (n = 48) and aged, hyperlipidemic LDLR-/-ApoB100/100 mice (n = 69) after the induction of acute hindlimb ischemia using contrast ultrasound, photoacoustic imaging and histological analyses, respectively. The capillary responses that led to successful post-ischemic muscle repair in C57Bl/6J mice included an early capillary dilation phase, preceding the return of arterial driving pressure, followed by an increase in capillary density that further supported satellite cell-induced muscle regeneration. Initial capillary enlargement was absent in the LDLR-/-ApoB100/100 mice with lifelong moderate hypercholesterolemia and led to an inability to recover arterial driving pressure, with a resulting increase in distal necrosis, chronic tissue damage and a delay in the overall recovery after ischemia. To conclude, this manuscript highlights, beyond arterial collateralization, the importance of the proper function of the capillary endothelium in post-ischemic recovery and displays how post-ischemic capillary dynamics associate beyond tissue blood flow to both hemoglobin oxygenation and tissue regeneration.


Subject(s)
Arteries , Ischemia , Animals , Mice , Endothelium, Vascular , Mice, Inbred C57BL , Muscles , Hindlimb
4.
Hum Gene Ther ; 32(13-14): 761-770, 2021 07.
Article in English | MEDLINE | ID: mdl-33371775

ABSTRACT

Biological bypass through induced angiogenesis by vascular endothelial growth factor D (VEGF-D) gene therapy (GT) is a new concept for the treatment of cardiac ischemia. Serotype 5 adenoviruses are used in the clinical trials for transferring the VEGF-D cDNA into the ischemic myocardium. However, the presence of replication-competent vectors in the adenovirus products is a widely recognized problem that may pose a potential safety risk to the treated patients. We compared three different VEGF-D GT production lots containing different levels of replication-competent adenoviruses (RCA) tested in 3 × 1010 viral particles (vp): <10 RCA (VEGF-D L-RCA1), 10-100 RCA (VEGF-D H-RCA2), and 100-200 RCA (VEGF-D H-RCA3), as measured by a novel droplet digital polymerase chain reaction (PCR) RCA assay in a preclinical rabbit model (n = 21). ß-galactosidase encoding nonclinical-grade preparation was used as a nonangiogenic control. Each preparation was injected into the right semimembranosus muscle using dose of 1 × 1011 vp. Efficacy of the products was tested by the combination of contrast pulse sequencing ultrasound and modified Miles assay as well as quantifying the total cross-sectional area of capillaries. Safety, immunogenicity, toxicity, biodistribution, and shedding were assessed by general histology, serial measurements of C-reactive protein, white blood cell count and body temperature as well as using quantitative real-time PCR with primers targeted to the VEGF-D and replication-permitting E1 sequences. We found no significant differences in the efficacy or safety between the study groups. Most importantly, no detectable presence of RCA-specific E1 sequence was found in any samples tested, indicating that no detectable vector replication took place in vivo. We conclude that relatively low levels of RCA in adenoviral GT products may not be as important major safety issue as previously anticipated.


Subject(s)
Adenoviruses, Human , Vascular Endothelial Growth Factor D , Adenoviridae/genetics , Adenoviridae/metabolism , Animals , Genetic Therapy , Genetic Vectors/genetics , Humans , Neovascularization, Pathologic , Rabbits , Tissue Distribution , Vascular Endothelial Growth Factor D/genetics , Vascular Endothelial Growth Factor D/metabolism
5.
Vasc Biol ; 1(1): 1-9, 2019.
Article in English | MEDLINE | ID: mdl-32923945

ABSTRACT

BACKGROUND: Prosthetic vascular grafts in humans characteristically lack confluent endothelialization regardless of the duration of implantation. Use of high-porosity grafts has been proposed as a way to induce endothelialization through transgraft capillarization, although early experiments failed to show increased healing in man. OBJECTIVES: We hypothesized that transduction of tissues around the prosthetic conduit with vectors encoding VEGF receptor-2 (VEGFR2) ligands would augment transinterstitial capillarization and induce luminal endothelialization of high-porosity ePTFE grafts. METHODS: Fifty-two NZW rabbits received 87 ePTFE uni- or bilateral end-to-end interposition grafts in carotid arteries. Rabbits were randomized to local therapy with adenoviruses encoding AdVEGF-A165, AdVEGF-A109 or control AdLacZ and analyzed at 6 and 28 days after surgery by contrast-enhanced ultrasound and histology. RESULTS: AdVEGF-A165 and AdVEGF-A109 dramatically increased perfusion in perigraft tissues at 6 days (14.2 ± 3.6 or 16.7 ± 2.6-fold increases, P < 0.05 and P < 0.01). At 28 days, the effect was no longer significantly higher than baseline. At 6 days, no luminal endothelialization was observed in any of the groups. At 28 days, AdVEGF-A109- and AdVEGF-A165-treated animals showed enhanced ingrowth of transinterstitial capillaries (66.0 ± 13.7% and 77.4 ± 15.7% of graft thickness vs 44.7 ± 24.4% in controls, P < 0.05) and improved luminal endothelialization (11.2 ± 26.3% and 11.4 ± 22.2%, AdVEGF-A109 and AdVEGF-A165 vs 0% in controls, P < 0.05). No increased stenosis was observed in the treatment groups as compared to LacZ controls. CONCLUSIONS: This study suggests that transient local overexpression of VEGFR2 ligands in the peri-implant tissues at the time of graft implantation is a novel strategy to increase endothelialization of high-porosity ePTFE vascular grafts and improve the patency of small-diameter vascular prostheses.

6.
NMR Biomed ; 31(5): e3909, 2018 05.
Article in English | MEDLINE | ID: mdl-29570882

ABSTRACT

The identification of areas with regenerative potential in ischemic tissues would allow the targeting of treatments supporting tissue recovery. The regeneration process involves the activation of several cellular and molecular responses which could be detected using magnetic resonance imaging (MRI). However, to date, magnetic resonance (MR) relaxation parameters have received little attention in the diagnosis and follow-up of limb ischemia. The purpose of this study was to evaluate the feasibility of different MRI relaxation and diffusion tensor imaging parameters in the detection of areas showing early signs of regeneration in ischemic mouse skeletal muscles. T2 and T1ρ relaxation time constants, together with TRAFFn , T1 and diffusion tensor imaging, were evaluated to differentiate areas of regeneration in a mouse hind limb ischemia model before and 0, 1, 4, 7, 14 and 30 days after ischemia. All the measured relaxation times were longer in the areas of early regeneration compared with normal muscle tissue. The relaxation times increased after ischemia in the ischemic muscles, reaching a maximum at 4-7 days after occlusion, coinciding with the appearance of early signs of regeneration. Fractional anisotropy decreased significantly (p < 0.05) on days 1-4, whereas mean diffusivity, λ1 and λ2 decreased later, starting at day 7 after ischemia compared with the pre-operational time point. The percentages of areas with different tissue morphologies were determined based on histological analysis of the ischemic muscle cross-sections, and correlations between the percentages obtained and different relaxation times were calculated. The highest correlation between relaxation times and histology was achieved with T2 , T1ρ and TRAFF4 (R2 = 0.96, R2 = 0.92 and R2 = 0.84, respectively, p < 0.01). Early regenerative changes were visible using T2 , T1ρ and TRAFF4 MR relaxation time constants in skeletal muscle after ischemia. These markers could potentially be used for the identification of targets for therapies supporting muscle regeneration after ischemic injury.


Subject(s)
Diffusion Tensor Imaging , Ischemia/diagnostic imaging , Muscle, Skeletal/blood supply , Muscle, Skeletal/pathology , Regeneration , Animals , Female , Ischemia/pathology , Mice , Muscle, Skeletal/diagnostic imaging , Time Factors
7.
Eur Heart J ; 38(18): 1365-1371, 2017 May 07.
Article in English | MEDLINE | ID: mdl-28073865

ABSTRACT

Chronic cardiovascular diseases are significant health problems. Although current treatment strategies have tremendously improved disease management, up to 30% of these patients cannot be successfully treated with current treatment approaches and new treatment strategies are clearly needed. Gene therapy and therapeutic vascular growth may provide a new treatment option for these patients. Several growth factors, like vascular endothelial growth factors, fibroblast growth factors and hepatocyte growth factor have been tested in clinical trials. However, apart from demonstration of increased vascularity, very few results with clinical significance have been obtained. Problems with gene transfer efficiency, short duration of transgene expression, selection of endpoints, and suboptimal patients for gene therapy have been recognized. Ongoing gene therapy trials have included improvements in study protocols, vector delivery and endpoints, addressing the identified problems. Better, targeted delivery systems and new, more optimal growth factors have been taken to clinical testing. Recent advances in these areas will be discussed and the concept of angiogenic therapy as a sole treatment is re-evaluated. A combination with regenerative therapies or standard revascularization operations might be needed to improve tissue function and clinical benefits.


Subject(s)
Angiogenesis Inducing Agents/therapeutic use , Cardiovascular Diseases/therapy , Genetic Therapy/methods , Clinical Trials as Topic , Forecasting , Gene Transfer Techniques , Genetic Vectors , Heart Failure/therapy , Humans , Myocardial Ischemia/therapy , Patient Selection , Peripheral Vascular Diseases/therapy , Vascular Endothelial Growth Factors/genetics
8.
Am J Physiol Heart Circ Physiol ; 307(8): H1226-32, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-25320333

ABSTRACT

Highly increased blood flow and vascularity after angiogenic gene therapy have raised concerns of shunting and hemangioma-like blood pool formation that might decrease effective perfusion and ruin the beneficial effects of the therapy. Contrast enhanced ultrasound is a promising noninvasive tool for studying skeletal muscle perfusion. The objectives of the present study were to test bolus and infusion administrations of ultrasound microbubble contrast media in imaging vascular growth in skeletal muscle and assess the functionality of vessels grown with angiogenic gene therapy. Contrast enhanced ultrasound was used to study changes in skeletal muscle perfusion in normal and gene-transduced rabbit hindlimbs 6 days after gene transfer. Adenoviral gene transfer of VEGF (10e(9)-10e(11) viral particles) or ß-galactosidase control gene (10e(11) viral particles) was done under anesthesia and induced up to 16-fold increases in relative tissue perfusion. Contrast intensity versus time curves were plotted and analyzed for contrast kinetics. Bolus administration of the contrast media was highly feasible in analyzing skeletal muscle blood flow and its kinetics. Maximal signal intensity of the bolus signal reflected relative changes in both blood flow and volume equally to the infusion method. Flow irregularities were detected after angiogenic gene therapy. In conclusion, bolus delivery of ultrasound contrast agent is highly feasible for the relative analysis of both quantity and quality of blood flow after angiogenic gene therapy. The kinetics of blood flow can and should be studied more extensively in both preclinical and clinical trials of angiogenic gene therapy since there is increasing evidence of flow irregularities in angiogenic vessels.


Subject(s)
Contrast Media/pharmacokinetics , Muscle, Skeletal/blood supply , Neovascularization, Physiologic , Ultrasonography/methods , Animals , Hindlimb/blood supply , Hindlimb/diagnostic imaging , Microbubbles , Muscle, Skeletal/diagnostic imaging , Rabbits , Regional Blood Flow
9.
Eur J Clin Invest ; 44(10): 989-99, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25208310

ABSTRACT

BACKGROUND: Cardiovascular patients suffer from reduced blood flow leading to ischaemia and impaired tissue metabolism. Unfortunately, an increasing group of elderly patients cannot be treated with current revascularization methods. Thus, new treatment strategies are urgently needed. Hypoxia-inducible factors (HIFs) upregulate the expression of angiogenic mediators together with genes involved in energy metabolism and recovery of ischaemic tissues. Especially, HIF-2α is a novel factor, and only limited information is available about its therapeutic potential. METHODS: Gene transfers with adenoviral HIF-1α and HIF-2α were performed into the mouse heart and rabbit ischaemic hindlimbs. Angiogenesis was evaluated by histology. Left ventricle function was analysed with echocardiography. Perfusion in rabbit skeletal muscles and energy recovery after electrical stimulation-induced exercise were measured with ultrasound and (31)P-magnetic resonance spectroscopy ((31)P-MRS), respectively. RESULTS: HIF-1α and HIF-2α gene transfers increased capillary size up to fivefold in myocardium and ischaemic skeletal muscles. Perfusion in skeletal muscles was increased by fourfold without oedema. Especially, AdHIF-1α enhanced the recovery of ischaemic muscles from electrical stimulation-induced energy depletion. Special characteristic of HIF-2α gene transfer was a strong capillary growth in muscle connective tissue and that HIF-2α gene transfer maintained left ventricle function. CONCLUSIONS: We conclude that both AdHIF-1α and AdHIF-2α gene transfers induced beneficial angiogenesis in vivo. Transient moderate increases in angiogenesis improved energy recovery after exercise in ischaemic muscles. This study shows for the first time that a moderate increase in angiogenesis is enough to improve tissue energy metabolism, which is potentially a very useful feature for cardiovascular gene therapy.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/pharmacology , Hypoxia-Inducible Factor 1, alpha Subunit/pharmacology , Muscle, Skeletal/metabolism , Neovascularization, Physiologic/drug effects , Animals , Capillaries/physiology , Coronary Vessels/physiology , Gene Expression/physiology , Gene Transfer Techniques , Genetic Therapy/methods , Hindlimb/blood supply , Ischemia/physiopathology , Ischemia/therapy , Mice, Inbred C57BL , Muscle, Skeletal/blood supply , Myocardium/metabolism , Rabbits
10.
ACS Nano ; 7(4): 3362-72, 2013 Apr 23.
Article in English | MEDLINE | ID: mdl-23451983

ABSTRACT

Herein, we report the use of biodegradable nanoparticles (NPs) containing perfluoro-1,5-crown ether (PFCE), a fluorine-based compound (NP170-PFCE) with the capacity to track cells in vivo by magnetic ressonance imaging (MRI) and efficiently release miRNA. NP170-PFCE complexed with miRNAs accumulate whitin the cell's endolysosomal compartment and interact with higher frequency with argonaute2 (Ago2) and GW182 proteins, which are involved in the biological action of miRNAs, than commercial complexes formed by commercial reagents and miRNA, which in turn accumulate in the cell cytoplasm. The release of miRNA132 (miR132) from the NPs increased 3-fold the survival of endothelial cells (ECs) transplanted in vivo and 3.5-fold the blood perfusion in ischemic limbs relatively to control.


Subject(s)
Ischemia/pathology , Ischemia/therapy , Magnetic Resonance Imaging/methods , MicroRNAs/administration & dosage , MicroRNAs/genetics , Nanocapsules/therapeutic use , Angiogenesis Inducing Agents/administration & dosage , Animals , Cell Survival/drug effects , Cell Tracking/methods , Cells, Cultured , Endothelial Cells/physiology , Hindlimb/blood supply , Humans , Mice , Nanocapsules/chemistry , Transfection/methods
11.
Dis Model Mech ; 6(2): 312-22, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23471910

ABSTRACT

Angiogenic therapy, which involves the use of an exogenous stimulus to promote blood vessel growth, is an attractive approach for the treatment of ischemic diseases. It has been shown in animal models that the stimulation of blood vessel growth leads to the growth of the whole vascular tree, improvement of ischemic tissue perfusion and improved muscle aerobic energy metabolism. However, very few positive results have been gained from Phase 2 and 3 clinical angiogenesis trials. Many reasons have been given for the failures of clinical trials, including poor transgene expression (in gene-therapy trials) and instability of the vessels induced by therapy. In this Review, we discuss the selection of preclinical models as one of the main reasons why clinical translation has been unsuccessful thus far. This issue has received little attention, but could have had dramatic implications on the expectations of clinical trials. We highlight crucial differences between human patients and animal models with regards to blood flow and pressure, as well as issues concerning the chronic nature of ischemic diseases in humans. We use these as examples to demonstrate why the results from preclinical trials might have overestimated the efficacy of angiogenic therapies developed to date. We also suggest ways in which currently available animal models of ischemic disease could be improved to better mimic human disease conditions, and offer advice on how to work with existing models to avoid overestimating the efficacy of new angiogenic therapies.


Subject(s)
Blood Vessels/growth & development , Ischemia/pathology , Neovascularization, Physiologic , Translational Research, Biomedical , Angiogenesis Inducing Agents/pharmacology , Angiogenesis Inducing Agents/therapeutic use , Animals , Blood Vessels/drug effects , Disease Models, Animal , Humans , Ischemia/drug therapy , Neovascularization, Physiologic/drug effects
12.
Circulation ; 127(4): 424-34, 2013 Jan 29.
Article in English | MEDLINE | ID: mdl-23357661

ABSTRACT

BACKGROUND: There is an unmet need for proangiogenic therapeutic molecules for the treatment of tissue ischemia in cardiovascular diseases. However, major inducers of angiogenesis such as vascular endothelial growth factor (VEGF/VEGF-A) have side effects that limit their therapeutic utility in vivo, especially at high concentrations. Angiopoietin-1 has been considered to be a blood vessel stabilization factor that can inhibit the intrinsic property of VEGF to promote vessel leakiness. In this study, we have designed and tested the angiogenic properties of chimeric molecules consisting of receptor-binding parts of VEGF and angiopoietin-1. We aimed at combining the activities of both factors into 1 molecule for easy delivery and expression in target tissues. METHODS AND RESULTS: The VEGF-angiopoietin-1 (VA1) chimeric protein bound to both VEGF receptor-2 and Tie2 and induced the activation of both receptors. Detailed analysis of VA1 versus VEGF revealed differences in the kinetics of VEGF receptor-2 activation and endocytosis, downstream kinase activation, and VE-cadherin internalization. The delivery of a VA1 transgene into mouse skeletal muscle led to increased blood flow and enhanced angiogenesis. VA1 was also very efficient in rescuing ischemic limb perfusion. However, VA1 induced less plasma protein leakage and myeloid inflammatory cell recruitment than VEGF. Furthermore, angioma-like structures associated with VEGF expression were not observed with VA1. CONCLUSIONS: The VEGF-angiopoietin-1 chimera is a potent angiogenic factor that triggers a novel mode of VEGF receptor-2 activation, promoting less vessel leakiness, less tissue inflammation, and better perfusion in ischemic muscle than VEGF. These properties of VA1 make it an attractive therapeutic tool.


Subject(s)
Angiopoietin-1/pharmacology , Genetic Therapy/methods , Ischemia/drug therapy , Neovascularization, Physiologic/physiology , Recombinant Fusion Proteins/pharmacology , Vascular Endothelial Growth Factor A/pharmacology , Adenoviridae/genetics , Angiopoietin-1/genetics , Angiopoietin-1/metabolism , Animals , Capillary Permeability/physiology , Cell Line, Tumor , Disease Models, Animal , Female , HEK293 Cells , Hindlimb/blood supply , Human Umbilical Vein Endothelial Cells , Humans , Ischemia/genetics , Leukemia, Myeloid , Mice , Mice, Inbred Strains , Muscle, Skeletal/blood supply , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, TIE-2 , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Signal Transduction/physiology , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-1/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
13.
Eur Heart J ; 32(13): 1664-72, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21138938

ABSTRACT

AIMS: Currently, it is still unclear which mechanisms drive metabolic benefits after angiogenic gene therapy. The side-effect profile of efficient angiogenic gene therapy is also currently incompletely understood. In this study, the effects of increasing doses of adenoviral (Ad) vascular endothelial growth factor-A (VEGF-A) were evaluated on vascular growth, metabolic benefits, and systemic side effects. METHODS AND RESULTS: Adenoviral vascular endothelial growth factor-A or AdLacZ control was injected intramuscularly (10(9)-10(11) vp/mL) or intra-arterially (5 × 10(11) vp/mL) into rabbit (n = 102) hindlimb muscles and examined 6 or 14 days later. Blood flow, tissue oedema, metabolic benefits, and the structure of angiogenic vessels were assessed using ultrasound imaging, modified Miles assay, arterial blood gas and metabolite analyses, and light and confocal microscopy, respectively. Safety analyses included cardiac ultrasound, electrocardiograms, and blood and tissue samples. Sprouting angiogenesis was already induced with low AdVEGF-A concentrations, whereas higher concentrations were needed to reach efficient capillary enlargement and increases in target muscle perfusion. Interestingly, metabolic benefits, such as improved aerobic energy metabolism and decreased metabolic acidosis during exercise, after AdVEGF-A administration were highly correlated to the level of capillary enlargement but not to sprouting angiogenesis. Several systemic dose-dependent side effects, including transient increases in liver, kidney, and pancreatic enzymes, and signs of cardiac effects were observed. CONCLUSION: Efficient capillary enlargement leading to significant increases in tissue perfusion is needed to gain metabolic benefits after angiogenic gene therapy. However, the risk of systemic side effects can increase as the efficiency of angiogenic gene therapy is improved. Importantly, the unstable wall structure of the newly formed vessels seems not to compromise the metabolic benefits.


Subject(s)
Capillaries/anatomy & histology , Genetic Therapy/methods , Muscle, Skeletal/blood supply , Neovascularization, Physiologic , Vascular Endothelial Growth Factor A/administration & dosage , Adenoviridae , Animals , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Gene Transfer Techniques , Genetic Therapy/adverse effects , Genetic Vectors , Hindlimb , Injections, Intramuscular , Lac Operon/genetics , Rabbits , Ultrasonography, Interventional , Vascular Endothelial Growth Factor A/adverse effects
14.
Circulation ; 122(17): 1725-33, 2010 Oct 26.
Article in English | MEDLINE | ID: mdl-20937974

ABSTRACT

BACKGROUND: Vascular endothelial growth factor-B (VEGF-B) binds to VEGF receptor-1 and neuropilin-1 and is abundantly expressed in the heart, skeletal muscle, and brown fat. The biological function of VEGF-B is incompletely understood. METHODS AND RESULTS: Unlike placenta growth factor, which binds to the same receptors, adeno-associated viral delivery of VEGF-B to mouse skeletal or heart muscle induced very little angiogenesis, vascular permeability, or inflammation. As previously reported for the VEGF-B(167) isoform, transgenic mice and rats expressing both isoforms of VEGF-B in the myocardium developed cardiac hypertrophy yet maintained systolic function. Deletion of the VEGF receptor-1 tyrosine kinase domain or the arterial endothelial Bmx tyrosine kinase inhibited hypertrophy, whereas loss of VEGF-B interaction with neuropilin-1 had no effect. Surprisingly, in rats, the heart-specific VEGF-B transgene induced impressive growth of the epicardial coronary vessels and their branches, with large arteries also seen deep inside the subendocardial myocardium. However, VEGF-B, unlike other VEGF family members, did not induce significant capillary angiogenesis, increased permeability, or inflammatory cell recruitment. CONCLUSIONS: VEGF-B appears to be a coronary growth factor in rats but not in mice. The signals for the VEGF-B-induced cardiac hypertrophy are mediated at least in part via the endothelium. Because cardiomyocyte damage in myocardial ischemia begins in the subendocardial myocardium, the VEGF-B-induced increased arterial supply to this area could have therapeutic potential in ischemic heart disease.


Subject(s)
Capillary Permeability/physiology , Coronary Vessels/growth & development , Inflammation/physiopathology , Neovascularization, Physiologic/physiology , Vascular Endothelial Growth Factor B/physiology , Adenoviridae/genetics , Animals , Cardiomegaly/physiopathology , Female , Humans , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Transgenic , Models, Animal , Muscle, Skeletal/blood supply , Myocardium , Neuropilin-1/physiology , Rats , Rats, Transgenic , Rats, Wistar , Vascular Endothelial Growth Factor B/genetics
15.
Integr Biol (Camb) ; 2(2-3): 102-12, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20473388

ABSTRACT

The process of growing new blood vessels through gene therapy may be difficult but is certainly possible. This review will discuss the most important factors determining the efficacy of angiogenic gene therapy.


Subject(s)
Blood Vessels/growth & development , Genetic Therapy/trends , Neovascularization, Physiologic/physiology , Tissue Engineering/trends , Animals , Humans
16.
Circ Res ; 105(6): 604-9, 2009 Sep 11.
Article in English | MEDLINE | ID: mdl-19696410

ABSTRACT

RATIONALE: We studied a possibility that shRNAs can lead to transcriptional gene activation at the promoter level via epigenetic mechanism. OBJECTIVE: The purpose of this study was to test the effects on vascular endothelial growth factor (VEGF-A) expression by promoter targeted small hairpin RNAs (shRNAs) in vitro and in experimental animals in vivo using stable local lentiviral gene transfer. METHODS AND RESULTS: One shRNA was identified which strongly increased VEGF-A expression in C166 endothelial cells at mRNA and protein level whereas another shRNA decreased VEGF-A expression. Quantitative chromatin immunoprecipitation analysis revealed that the repressing shRNA caused epigenetic changes, which increased nucleosome density within the promoter and transcription start site and led to repression of VEGF-A expression. Epigenetic changes caused by the activating shRNA were opposite to those caused by the repressing shRNA. These results were confirmed in vivo in an ischemic mouse hindlimb model after local gene transfer where VEGF-A upregulation achieved by promoter-targeted shRNA increased vascularity and blood flow. CONCLUSIONS: We show that lentivirus-mediated delivery of shRNA molecules targeted to specific regions in the mVEGF-A promoter either induce or repress VEGF-A expression via epigenetic modulation. Thus, we describe a new approach of gene therapy, epigenetherapy, based on an epigenetic mechanism at the promoter level. Controlling transcription through manipulation of specific epigenetic marks provides a novel approach for the treatment of several diseases.


Subject(s)
Epigenesis, Genetic , Genetic Therapy/methods , Hindlimb/blood supply , Ischemia/therapy , Lentivirus , Promoter Regions, Genetic , RNA/biosynthesis , Vascular Endothelial Growth Factor A/genetics , Animals , Cell Line , Endothelial Cells/metabolism , Ischemia/genetics , Mice , RNA/genetics , Transcription, Genetic , Vascular Endothelial Growth Factor A/metabolism
17.
Circ Res ; 104(11): 1302-12, 2009 Jun 05.
Article in English | MEDLINE | ID: mdl-19443835

ABSTRACT

The therapeutic potential of vascular endothelial growth factor (VEGF)-C and VEGF-D in skeletal muscle has been of considerable interest as these factors have both angiogenic and lymphangiogenic activities. Previous studies have mainly used adenoviral gene delivery for short-term expression of VEGF-C and VEGF-D in pig, rabbit, and mouse skeletal muscles. Here we have used the activated mature forms of VEGF-C and VEGF-D expressed via recombinant adeno-associated virus (rAAV), which provides stable, long-lasting transgene expression in various tissues including skeletal muscle. Mouse tibialis anterior muscle was transduced with rAAV encoding human or mouse VEGF-C or VEGF-D. Two weeks later, immunohistochemical analysis showed increased numbers of both blood and lymph vessels, and Doppler ultrasound analysis indicated increased blood vessel perfusion. The lymphatic vessels further increased at the 4-week time point were functional, as shown by FITC-lectin uptake and transport. Furthermore, receptor activation and arteriogenic activity were increased by an alanine substitution mutant of human VEGF-C (C137A) having an increased dimer stability and by a chimeric CAC growth factor that contained the VEGF receptor-binding domain flanked by VEGF-C propeptides, but only the latter promoted significantly more blood vessel perfusion when compared to the other growth factors studied. We conclude that long-term expression of VEGF-C and VEGF-D in skeletal muscle results in the generation of new functional blood and lymphatic vessels. The therapeutic value of intramuscular lymph vessels in draining tissue edema and lymphedema can now be evaluated using this model system.


Subject(s)
Blood Vessels/physiology , Heart/physiology , Lymphatic Vessels/physiology , Muscle, Skeletal/physiology , Vascular Endothelial Growth Factor B/physiology , Vascular Endothelial Growth Factor C/physiology , Animals , Dimerization , Drug Stability , Humans , Mice , Mice, Transgenic , Muscle, Skeletal/blood supply , Mutation , Polymorphism, Single Nucleotide , Recombinant Proteins/metabolism , Vascular Endothelial Growth Factor B/genetics , Vascular Endothelial Growth Factor C/genetics , Vascular Endothelial Growth Factor Receptor-2/physiology
18.
Circulation ; 119(6): 845-56, 2009 Feb 17.
Article in English | MEDLINE | ID: mdl-19188502

ABSTRACT

BACKGROUND: New revascularization therapies are urgently needed for patients with severe coronary heart disease who lack conventional treatment options. METHODS AND RESULTS: We describe a new proangiogenic approach for these no-option patients using adenoviral (Ad) intramyocardial vascular endothelial growth factor (VEGF)-B186 gene transfer, which induces myocardium-specific angiogenesis and arteriogenesis in pigs and rabbits. After acute infarction, AdVEGF-B186 increased blood vessel area, perfusion, ejection fraction, and collateral artery formation and induced changes toward an ischemia-resistant myocardial phenotype. Soluble VEGF receptor-1 and soluble neuropilin receptor-1 reduced the effects of AdVEGF-B186, whereas neither soluble VEGF receptor-2 nor inhibition of nitric oxide production had this result. The effects of AdVEGF-B186 involved activation of neuropilin receptor-1, which is highly expressed in the myocardium, via recruitment of G-protein-alpha interacting protein, terminus C (GIPC) and upregulation of G-protein-alpha interacting protein. AdVEGF-B186 also induced an antiapoptotic gene expression profile in cardiomyocytes and had metabolic effects by inducing expression of fatty acid transport protein-4 and lipid and glycogen accumulation in the myocardium. CONCLUSIONS: VEGF-B186 displayed strikingly distinct effects compared with other VEGFs. These effects may be mediated at least in part via a G-protein signaling pathway. Tissue-specificity, high efficiency in ischemic myocardium, and induction of arteriogenesis and antiapoptotic and metabolic effects make AdVEGF-B186 a promising candidate for the treatment of myocardial ischemia.


Subject(s)
Arteries/drug effects , Myocardial Ischemia/therapy , Neovascularization, Physiologic/drug effects , Neuropilin-1/metabolism , Vascular Endothelial Growth Factor B/administration & dosage , Vascular Endothelial Growth Factor Receptor-1/metabolism , Animals , Arteries/growth & development , Genetic Therapy/methods , Genetic Vectors , Myocardial Infarction/therapy , Myocardial Reperfusion Injury/prevention & control , Organ Specificity , Rabbits , Swine
19.
Circ Res ; 103(10): 1092-9, 2008 Nov 07.
Article in English | MEDLINE | ID: mdl-18832750

ABSTRACT

Vessel stabilization and the inhibition of side effects such as tissue edema are essential in angiogenic gene therapy. Thus, combination gene transfers stimulating both endothelial cell and pericyte proliferation have become of interest. However, there is currently little data to support combination gene transfer in large animal models. In this study, we evaluated the potential advantages of such a strategy by combining the transfer of adenoviral (Ad) vascular endothelial growth factor (VEGF)-A and platelet-derived growth factor (PDGF)-B into rabbit hindlimb skeletal muscle. AdLacZ alone or in combination with AdVEGF-A were used as controls. Contrast-enhanced ultrasound, modified Miles assay, and immunohistology were used to quantify perfusion, vascular permeability, and capillary size, respectively. Confocal microscopy was used in the assessment of pericyte-coverage. The transfer of AdPDGF-B alone and in combination with AdVEGF-A induced prominent proliferation of alpha-smooth muscle actin-, CD31-, RAM11-, HAM56-, and VEGF- positive cells. Although, pericyte recruitment to angiogenic vessels was not improved, combination gene transfer induced a longer-lasting increase in perfusion in both intact and ischemic muscles than AdVEGF-A gene transfer alone. In conclusion, intramuscular delivery of AdVEGF-A and AdPDGF-B, combined, resulted in a prolonged angiogenic response. However, the effects were most likely mediated via paracrine mechanisms rather than an increase in vascular pericyte coverage.


Subject(s)
Cell Movement , Genetic Therapy/methods , Neovascularization, Physiologic , Paracrine Communication , Pericytes/metabolism , Proto-Oncogene Proteins c-sis/biosynthesis , Vascular Endothelial Growth Factor A/biosynthesis , Adenoviridae , Animals , Capillary Permeability/genetics , Cell Movement/genetics , Cell Proliferation , Humans , Muscle, Skeletal/cytology , Muscle, Skeletal/diagnostic imaging , Muscle, Skeletal/metabolism , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Neovascularization, Physiologic/genetics , Paracrine Communication/genetics , Pericytes/cytology , Platelet Endothelial Cell Adhesion Molecule-1/biosynthesis , Proto-Oncogene Proteins c-sis/genetics , Rabbits , Time Factors , Ultrasonography , Vascular Endothelial Growth Factor A/genetics
20.
Cardiovasc Res ; 80(2): 263-70, 2008 Nov 01.
Article in English | MEDLINE | ID: mdl-18647755

ABSTRACT

AIMS: We investigated the effects of angiogenic gene therapy with adenoviral placental growth factor(131) (AdPlGF) on aerobic capacity and exercise tolerance in a rabbit hindlimb ischaemia model. We also assessed whether strong angiogenic changes such as capillary arterialization and formation of artery-venous shunts compromise oxygen transport to target tissues resulting in suboptimal therapeutic efficacy. METHODS AND RESULTS: Hindlimb ischaemia was surgically induced in New Zealand White rabbits (n = 20) that a day later received intramuscular (i.m.) AdPlGF or AdLacZ (3 x 10(11)vp) gene transfer (GT). Corresponding GTs were also done in healthy non-ischaemic rabbits (n = 10). Muscle energy metabolism and skeletal muscle perfusion were studied non-invasively before GT and at 6 and 28 days using (31)P-magnetic resonance spectroscopy and contrast pulse sequence ultrasound, respectively. Oedema was quantified using modified Miles assay at sacrifice. AdPlGF increased perfusion 7.8-fold and improved aerobic capacity of ischaemic limbs 45% compared with AdLacZ controls (P < 0.05) at 6 days. In non-ischaemic limbs, strong angiogenic response to GT, including capillary arterialization and acute oedema, did not impair muscle energy metabolism. CONCLUSION: This study shows that proangiogenic gene therapy can significantly improve performance of ischaemic limbs and supports the concept of therapeutic angiogenesis for the treatment of patients with ischaemia.


Subject(s)
Exercise Tolerance , Genetic Therapy/methods , Ischemia/therapy , Muscle, Skeletal/blood supply , Neovascularization, Physiologic , Pregnancy Proteins/biosynthesis , Acidosis/metabolism , Acidosis/physiopathology , Acidosis/prevention & control , Adenoviridae/genetics , Animals , Disease Models, Animal , Edema/etiology , Edema/metabolism , Edema/physiopathology , Energy Metabolism , Genetic Therapy/adverse effects , Genetic Vectors , Hindlimb , Ischemia/genetics , Ischemia/metabolism , Ischemia/physiopathology , Magnetic Resonance Spectroscopy , Mice , Placenta Growth Factor , Pregnancy Proteins/genetics , Rabbits , Regional Blood Flow , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...