Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Type of study
Language
Publication year range
1.
Circulation ; 110(12): 1638-44, 2004 Sep 21.
Article in English | MEDLINE | ID: mdl-15364809

ABSTRACT

BACKGROUND: Angiogenesis gene therapy with human tissue kallikrein (hTK) has shown promise for ischemic disease. The present study was undertaken to (1) assess an optimal gene transfer modality, (2) clarify hTK angiogenic pathways, and (3) discount possible side effects. METHODS AND RESULTS: The hTK gene was transferred to murine adductors by increasing doses of an adenovirus (Ad.hTK). Heterologous protein production was evaluated by ELISA and immunohistochemistry. Structural and functional characteristics of hTK-induced neovascularization were assessed. Muscular endothelial nitric oxide synthase (eNOS) and vascular endothelial growth factor (VEGF)-A mRNA and protein content were evaluated by real-time polymerase chain reaction and Western blotting. The ability of hTK to phosphorylate-activate Akt/protein kinase B (Akt-B) and VEGF receptor 2 (VEGF-R2) was also determined. Implication of the aforementioned mechanisms in Ad.hTK-induced neovascularization was challenged by blocking Akt-B with a dominant-negative Akt construct; NOS with N(G)-nitro-L-arginine methyl ester; and VEGF-A with neutralizing antibody, VEGF-R2 antagonist, or Ad carrying soluble VEGF-R1 gene. We found that 10(7) PFU Ad.hTK led to peak increases in capillary and arteriole density. Newly developed arterioles persisted for up to 8 weeks. Ad.hTK did not change microvascular permeability. Ad.hTK upregulated eNOS mRNA and protein and activated Akt-B through Ser-473 phosphorylation. Inhibitory studies documented that these biochemical events were instrumental to Ad.hTK-induced neovascularization. In contrast, Ad.hTK neither affected VEGF-A and VEGF-R2 levels nor increased VEGF-R2 phosphorylation. Consistently, Ad.hTK-induced neovascularization was not disturbed by any of the different approaches used to block VEGF-A. CONCLUSIONS: Our findings provide new information on the pathway involved in hTK-induced neoangiogenesis and represent an advancement toward clinical applications with Ad.hTK.


Subject(s)
Muscle, Skeletal/blood supply , Neovascularization, Physiologic/drug effects , Nitric Oxide Synthase/physiology , Protein Serine-Threonine Kinases/physiology , Proto-Oncogene Proteins/physiology , Tissue Kallikreins/physiology , Adenoviridae/genetics , Animals , Blotting, Western , Cell Division , Endothelial Cells/cytology , Endothelium, Vascular/cytology , Enzyme Induction/drug effects , Exudates and Transudates , Genes, Dominant , Genetic Vectors/pharmacology , Humans , Mice , Microcirculation , Muscle, Skeletal/metabolism , NG-Nitroarginine Methyl Ester/pharmacology , Neovascularization, Physiologic/genetics , Nitric Oxide Synthase/biosynthesis , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type II , Nitric Oxide Synthase Type III , Phosphorylation , Phosphoserine/analysis , Polymerase Chain Reaction , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-akt , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Tissue Kallikreins/genetics , Transduction, Genetic , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/analysis
2.
Diabetes ; 53(4): 1096-103, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15047627

ABSTRACT

Diabetes macro- and microvascular disease causes tissue hypoperfusion. This deficit, together with a failure to mount an adequate angiogenic response, might explain why vascular occlusion evolves more severely among diabetic patients. The present study investigated whether prophylactic gene therapy with human tissue kallikrein (hTK) may protect diabetic limbs from the consequences of supervening ischemia. Vehicle (saline) or an adenovirus carrying the gene for either hTK (Ad.hTK) or luciferase (Ad.Luc) was injected into left adductor muscles of streptozotocin-induced type 1 diabetic mice 2 weeks before operative occlusion of the ipsilateral femoral artery. Saline-injected nondiabetic mice served as controls. Hindlimb blood flow recovery was analyzed sequentially over the 2 weeks after ischemia induction. At necroscopy, microvessel density and endothelial cell proliferation and apoptosis were quantified in skeletal muscles. We found that limb perfusion recovery of saline-injected type 1 diabetic mice is delayed because of insufficient reparative neovascularization and excessive activation of endothelial cell apoptosis. By contrast, prophylactic Ad.hTK renewed the ability to mount an appropriate neovascularization response to ischemia, suppressed apoptosis, and upregulated endothelial nitric oxide synthase expression. Ultimately, correction of diabetic endotheliopathy by Ad.hTK allowed proper perfusion recovery as seen in nondiabetic mice. These discoveries disclose new therapeutic options for the treatment of diabetic complications.


Subject(s)
Diabetes Mellitus, Type 1/therapy , Diabetic Angiopathies/therapy , Genetic Therapy/methods , Tissue Kallikreins/genetics , Animals , Arterioles/drug effects , Arterioles/growth & development , Diabetes Mellitus, Experimental/therapy , Extremities/blood supply , Humans , Ischemia/etiology , Ischemia/therapy , Luciferases/genetics , Male , Mice , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type II , Nitric Oxide Synthase Type III , Reference Values , Time Factors
3.
Circulation ; 106(17): 2257-62, 2002 Oct 22.
Article in English | MEDLINE | ID: mdl-12390957

ABSTRACT

BACKGROUND: The neurotrophin nerve growth factor (NGF) regulates neuron survival and differentiation. Implication in neovascularization is supported by statement of NGF and its high-affinity receptor at vascular level and by NGF property of stimulating vascular endothelial cell proliferation. The present study investigated the involvement of endogenous NGF in spontaneous reparative response to ischemia. Mechanisms and therapeutic potential of NGF-induced neovascularization were examined. METHODS AND RESULTS: Unilateral limb ischemia was produced in CD1 mice by femoral artery resection. By ELISA and immunohistochemistry, we documented that statement of NGF and its high-affinity receptor is upregulated in ischemic muscles. The functional relevance of this phenomenon was assessed by means of NGF-neutralizing antibody. Chronic NGF blockade abrogated the spontaneous capillarization response to ischemia and augmented myocyte apoptosis. Then we tested whether NGF administration may exert curative effects. Repeated NGF injection into ischemic adductors increased capillary and arteriole density, reduced endothelial cell and myofiber apoptosis, and accelerated perfusion recovery, without altering systemic hemodynamics. In normoperfused muscles, NFG-induced capillarization was blocked by vascular endothelial growth factor-neutralizing antibodies, dominant-negative Akt, or NO synthase inhibition. CONCLUSIONS: These results indicate that NGF plays a functional role in reparative neovascularization. Furthermore, supplementation of the growth factor promotes angiogenesis through a vascular endothelial growth factor-Akt-NO-mediated mechanism. In the setting of ischemia, potentiation of NGF pathway stimulates angiogenesis and arteriogenesis, thereby accelerating hemodynamic recovery. NGF might be envisaged as a utilitarian target for the treatment of ischemic vascular disease.


Subject(s)
Arteries/growth & development , Ischemia/blood , Neovascularization, Physiologic , Nerve Growth Factor/physiology , Animals , Arteries/drug effects , Arterioles/drug effects , Arterioles/growth & development , Capillaries/drug effects , Capillaries/growth & development , Hemodynamics/drug effects , Hindlimb/blood supply , Ischemia/metabolism , Ischemia/physiopathology , Male , Mice , Muscle, Skeletal/blood supply , Muscle, Skeletal/metabolism , Neovascularization, Physiologic/drug effects , Nerve Growth Factor/antagonists & inhibitors , Nerve Growth Factor/pharmacology , Receptors, Nerve Growth Factor/metabolism , Up-Regulation
4.
Circ Res ; 91(4): 346-52, 2002 Aug 23.
Article in English | MEDLINE | ID: mdl-12193468

ABSTRACT

Proteinase-activated receptors (PAR-2) are expressed by the cardiovascular system and mediate vasodilation, plasma protein extravasation, and endothelial cell proliferation, all regarded as essential steps for neovascularization. We investigated the angiogenic action of PAR-2 signaling in vivo. The effect of the PAR-2 activating peptide (PAR-2AP, SLIGRL-NH2) was assessed in the absence of ischemia, and the therapeutic potential of PAR-2AP and the PAR-2 agonist trypsin (at 300 and 1.5 nmol IM daily for 21 days, respectively) was also tested in mice subjected to unilateral limb ischemia. PAR-2AP increased capillarity in normoperfused adductor skeletal muscles, whereas neither the vehicle of the PAR2-AP nor the PAR-2 reverse peptide (PAR-2RP, LRGILS-NH2) did produce any effect. In addition, both PAR-2AP and trypsin enhanced reparative angiogenic response to limb ischemia, an effect that was not produced by PAR-2RP or the vehicle of PAR-2 agonists. Potentiation of reparative angiogenesis by PAR-2AP or trypsin resulted in an accelerated hemodynamic recovery and enhanced limb salvage. In conclusions, our study is the first to demonstrate the angiogenic potential of PAR-2 stimulation in vivo. If similar effects occur in humans, PAR-2AP agonists could have some therapeutic potential for the treatment of tissue ischemia.


Subject(s)
Hindlimb/blood supply , Ischemia/physiopathology , Neovascularization, Physiologic , Receptors, Thrombin/metabolism , Animals , Capillaries/drug effects , Capillaries/pathology , Capillaries/physiopathology , Disease Models, Animal , Gene Expression Regulation/drug effects , Hemodynamics/drug effects , Hemodynamics/physiology , Injections, Intramuscular , Ischemia/drug therapy , Ischemia/pathology , Laser-Doppler Flowmetry , Male , Mice , Muscle, Skeletal/blood supply , Muscle, Skeletal/drug effects , Muscle, Skeletal/pathology , Neovascularization, Physiologic/drug effects , Neovascularization, Physiologic/physiology , Oligopeptides/pharmacology , Receptor, PAR-2 , Receptors, Thrombin/agonists , Receptors, Thrombin/genetics , Recovery of Function/drug effects , Regional Blood Flow/drug effects , Up-Regulation
5.
Circulation ; 106(8): 993-9, 2002 Aug 20.
Article in English | MEDLINE | ID: mdl-12186806

ABSTRACT

BACKGROUND: Microvascular insufficiency represents a major cause of end-organ failure among diabetics. METHODS AND RESULTS: In streptozotocin-induced diabetic mice, we evaluated the potential of human tissue kallikrein (hTK) gene as a sole therapy against peripheral microangiopathy. Local delivery of hTK gene halted the progression of microvascular rarefaction in hindlimb skeletal muscle by inhibiting apoptosis, thus ensuring an improved hemodynamic recovery in case of supervening vascular occlusion. The curative action of hTK did not necessitate insulin supplementation. Application of gene therapy at a stage of established microangiopathy stimulated vascular regeneration. CONCLUSIONS: Our studies indicate that hTK may represent a useful tool for the treatment of microvascular complications in diabetics.


Subject(s)
Diabetes Mellitus, Experimental/complications , Diabetic Angiopathies/prevention & control , Genetic Therapy , Ischemia/prevention & control , Tissue Kallikreins/genetics , Animals , Apoptosis , Combined Modality Therapy , Diabetic Angiopathies/etiology , Diabetic Angiopathies/pathology , Hindlimb/blood supply , Humans , Hypoglycemic Agents/therapeutic use , Insulin/therapeutic use , Ischemia/etiology , Ischemia/pathology , Kinetics , Male , Mice , Microcirculation/drug effects , Microcirculation/pathology , Muscle, Skeletal/blood supply , Muscle, Skeletal/chemistry , Muscle, Skeletal/pathology , Nucleotides, Cyclic/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...