Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
J Biomed Mater Res A ; 108(9): 1881-1889, 2020 09.
Article in English | MEDLINE | ID: mdl-32314537

ABSTRACT

Restoring blood supply is an effective way for the therapy of myocardial infarction (MI). It was reported a specific angiogenic peptide (VMP) derived from vascular endothelial growth factor (VEGF) could activate its receptor to mimic the biological activity of VEGF. In this study, in order to improve the local concentration in infarction region, a collagen-binding domain was synthesized with VMP to construct collagen binding domain (CBD)-VMP peptides. The fused CBD-VMP could bind specifically to collagen which was rich in cardiac extracellular matrix (c-ECM), without impacting the biological activity of VMP peptides. When the CBD-VMP peptides loaded on collagen scaffold and implanted into the rats subcutaneously, significant vascularization was observed. Then, CBD-VMP peptides binding with injectable c-ECM injected into the MI rat by intramuscular administration, significant blood vessels regeneration, and decrease of cell apoptosis were observed, that corelated with the recovery of cardiac function. It might be an alternative promising strategy for the clinical application of MI.


Subject(s)
Angiogenic Proteins/therapeutic use , Collagen/therapeutic use , Myocardial Infarction/drug therapy , Angiogenic Proteins/administration & dosage , Animals , Collagen/administration & dosage , Male , Myocardial Infarction/pathology , Neovascularization, Physiologic/drug effects , Rats , Rats, Sprague-Dawley
2.
J Biomed Mater Res A ; 107(12): 2764-2773, 2019 12.
Article in English | MEDLINE | ID: mdl-31408258

ABSTRACT

Angiogenesis plays a pivotal role in tissue regeneration following bone-grafting procedures; however, nonautogenous graft materials typically lack critical angiogenic growth factors. While much research has focused on modifying grafts with angiogenic factors, controlled delivery of these molecules remains a challenge. The current study describes a method for sustained delivery of an angiogenic peptide from hydroxyapatite (HA), a common alloplast material. Specifically, VEGF-derived "QK" peptides were synthesized with polyglutamate domains containing varying numbers of glutamates. The rate of peptide release from HA inversely correlated with glutamate number, with diglutamate-QK (E2-QK) released first, followed by tetraglutamate-QK (E4-QK), and finally, heptaglutamate-QK (E7-QK). By coating HA with a mixture of these peptides, termed, PGM-QK (polyglutamate-modified mixture), sequential peptide release was achieved, enabling gradient QK delivery. To evaluate bioactivity, HA disks were coated with PGM-QK and then placed in fresh media for 6 days. Media containing the released peptides was collected at varying time intervals and placed on human umbilical vein endothelial cells (HUVECs). Cells were evaluated for activation of angiogenic signaling pathways (ERK and Akt) and cell migration. Results showed that QK peptides were continuously released over the 6-day interval, and maintained their capacity to activate HUVECs. These findings point to a new approach for gradient delivery of an angiogenic stimulus.


Subject(s)
Angiogenesis Inducing Agents/administration & dosage , Angiogenic Proteins/administration & dosage , Bone Substitutes/chemistry , Delayed-Action Preparations/chemistry , Durapatite/chemistry , Polyglutamic Acid/administration & dosage , Angiogenesis Inducing Agents/chemistry , Angiogenesis Inducing Agents/pharmacology , Angiogenic Proteins/chemistry , Angiogenic Proteins/pharmacology , Drug Liberation , Human Umbilical Vein Endothelial Cells , Humans , Neovascularization, Physiologic/drug effects , Polyglutamic Acid/chemistry , Polyglutamic Acid/pharmacology , Vascular Endothelial Growth Factor A/administration & dosage , Vascular Endothelial Growth Factor A/chemistry , Vascular Endothelial Growth Factor A/pharmacology
3.
Circulation ; 136(19): 1809-1823, 2017 Nov 07.
Article in English | MEDLINE | ID: mdl-28931551

ABSTRACT

BACKGROUND: Clinical trials of bone marrow cell-based therapies after acute myocardial infarction (MI) have produced mostly neutral results. Treatment with specific bone marrow cell-derived secreted proteins may provide an alternative biological approach to improving tissue repair and heart function after MI. We recently performed a bioinformatic secretome analysis in bone marrow cells from patients with acute MI and discovered a poorly characterized secreted protein, EMC10 (endoplasmic reticulum membrane protein complex subunit 10), showing activity in an angiogenic screen. METHODS: We investigated the angiogenic potential of EMC10 and its mouse homolog (Emc10) in cultured endothelial cells and infarcted heart explants. We defined the cellular sources and function of Emc10 after MI using wild-type, Emc10-deficient, and Emc10 bone marrow-chimeric mice subjected to transient coronary artery ligation. Furthermore, we explored the therapeutic potential of recombinant Emc10 delivered by osmotic minipumps after MI in heart failure-prone FVB/N mice. RESULTS: Emc10 signaled through small GTPases, p21-activated kinase, and the p38 mitogen-activated protein kinase (MAPK)-MAPK-activated protein kinase 2 (MK2) pathway to promote actin polymerization and endothelial cell migration. Confirming the importance of these signaling events in the context of acute MI, Emc10 stimulated endothelial cell outgrowth from infarcted mouse heart explants via p38 MAPK-MK2. Emc10 protein abundance was increased in the infarcted region of the left ventricle and in the circulation of wild-type mice after MI. Emc10 expression was also increased in left ventricular tissue samples from patients with acute MI. Bone marrow-derived monocytes and macrophages were the predominant sources of Emc10 in the infarcted murine heart. Emc10 KO mice showed no cardiovascular phenotype at baseline. After MI, however, capillarization of the infarct border zone was impaired in KO mice, and the animals developed larger infarct scars and more pronounced left ventricular remodeling compared with wild-type mice. Transplanting KO mice with wild-type bone marrow cells rescued the angiogenic defect and ameliorated left ventricular remodeling. Treating FVB/N mice with recombinant Emc10 enhanced infarct border-zone capillarization and exerted a sustained beneficial effect on left ventricular remodeling. CONCLUSIONS: We have identified Emc10 as a previously unknown angiogenic growth factor that is produced by bone marrow-derived monocytes and macrophages as part of an endogenous adaptive response that can be enhanced therapeutically to repair the heart after MI.


Subject(s)
Angiogenic Proteins/metabolism , Bone Marrow Cells/metabolism , Membrane Proteins/metabolism , Myocardial Infarction/metabolism , Myocardium/metabolism , Neovascularization, Physiologic , Wound Healing , Angiogenic Proteins/administration & dosage , Angiogenic Proteins/deficiency , Angiogenic Proteins/genetics , Animals , Bone Marrow Transplantation , Cells, Cultured , Disease Models, Animal , Endothelial Cells/metabolism , Genotype , Intracellular Signaling Peptides and Proteins/metabolism , Macrophages/metabolism , Membrane Proteins/administration & dosage , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice, Inbred C57BL , Mice, Knockout , Monocytes/metabolism , Monomeric GTP-Binding Proteins/metabolism , Myocardial Infarction/drug therapy , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocardium/pathology , Neovascularization, Physiologic/drug effects , Phenotype , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Time Factors , Wound Healing/drug effects , p21-Activated Kinases/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
4.
J Am Heart Assoc ; 6(6)2017 Jun 25.
Article in English | MEDLINE | ID: mdl-28649088

ABSTRACT

BACKGROUND: Despite recent improvements in angioplasty and placement of drug-eluting stents in treatment of atherosclerosis, restenosis and in-stent thrombosis impede treatment efficacy and cause numerous deaths. Research efforts are needed to identify new molecular targets for blocking restenosis. We aim to establish angiogenic factor AGGF1 (angiogenic factor with G patch and FHA domains 1) as a novel target for blocking neointimal formation and restenosis after vascular injury. METHODS AND RESULTS: AGGF1 shows strong expression in carotid arteries; however, its expression is markedly decreased in arteries after vascular injury. AGGF1+/- mice show increased neointimal formation accompanied with increased proliferation of vascular smooth muscle cells (VSMCs) in carotid arteries after vascular injury. Importantly, AGGF1 protein therapy blocks neointimal formation after vascular injury by inhibiting the proliferation and promoting phenotypic switching of VSMCs to the contractile phenotype in mice in vivo. In vitro, AGGF1 significantly inhibits VSMCs proliferation and decreases the cell numbers at the S phase. AGGF1 also blocks platelet-derived growth factor-BB-induced proliferation, migration of VSMCs, increases expression of cyclin D, and decreases expression of p21 and p27. AGGF1 inhibits phenotypic switching of VSMCs to the synthetic phenotype by countering the inhibitory effect of platelet-derived growth factor-BB on SRF expression and the formation of the myocardin/SRF/CArG-box complex involved in activation of VSMCs markers. Finally, we show that AGGF1 inhibits platelet-derived growth factor-BB-induced phosphorylation of MEK1/2, ERK1/2, and Elk phosphorylation involved in the phenotypic switching of VSMCs, and that overexpression of Elk abolishes the effect of AGGF1. CONCLUSIONS: AGGF1 protein therapy is effective in blocking neointimal formation after vascular injury by regulating a novel AGGF1-MEK1/2-ERK1/2-Elk-myocardin-SRF/p27 signaling pathway.


Subject(s)
Angiogenic Proteins/administration & dosage , Carotid Artery Injuries/prevention & control , Carotid Stenosis/prevention & control , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Neointima , Angiogenic Proteins/deficiency , Angiogenic Proteins/genetics , Animals , Carotid Artery Injuries/genetics , Carotid Artery Injuries/metabolism , Carotid Artery Injuries/pathology , Carotid Artery, Common/drug effects , Carotid Artery, Common/metabolism , Carotid Artery, Common/pathology , Carotid Stenosis/genetics , Carotid Stenosis/metabolism , Carotid Stenosis/pathology , Cell Line , Cell Movement/drug effects , Cell Plasticity/drug effects , Cell Proliferation/drug effects , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation , Male , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinase Kinases/metabolism , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Nuclear Proteins/metabolism , Phenotype , Phosphorylation , RNA Interference , Serum Response Factor/metabolism , Signal Transduction/drug effects , Ternary Complex Factors/metabolism , Trans-Activators/metabolism , Transfection
5.
Biotechnol Bioeng ; 114(10): 2379-2389, 2017 10.
Article in English | MEDLINE | ID: mdl-28574594

ABSTRACT

In the last decade, numerous growth factors and biomaterials have been explored for the treatment of myocardial infarction (MI). While pre-clinical studies have demonstrated promising results, clinical trials have been disappointing and inconsistent, likely due to poor translatability. In the present study, we investigate a potential myocardial regenerative therapy consisting of a protein-engineered dimeric fragment of hepatocyte growth factor (HGFdf) encapsulated in a shear-thinning, self-healing, bioengineered hydrogel (SHIELD). We hypothesized that SHIELD would facilitate targeted, sustained intramyocardial delivery of HGFdf thereby attenuating myocardial injury and post-infarction remodeling. Adult male Wistar rats (n = 45) underwent sham surgery or induction of MI followed by injection of phosphate buffered saline (PBS), 10 µg HGFdf alone, SHIELD alone, or SHIELD encapsulating 10 µg HGFdf. Ventricular function, infarct size, and angiogenic response were assessed 4 weeks post-infarction. Treatment with SHIELD + HGFdf significantly reduced infarct size and increased both ejection fraction and borderzone arteriole density compared to the controls. Thus, sustained delivery of HGFdf via SHIELD limits post-infarction adverse ventricular remodeling by increasing angiogenesis and reducing fibrosis. Encapsulation of HGFdf in SHIELD improves clinical translatability by enabling minimally-invasive delivery and subsequent retention and sustained administration of this novel, potent angiogenic protein analog. Biotechnol. Bioeng. 2017;114: 2379-2389. © 2017 Wiley Periodicals, Inc.


Subject(s)
Delayed-Action Preparations/administration & dosage , Hepatocyte Growth Factor/administration & dosage , Hydrogels/chemistry , Myocardial Infarction/drug therapy , Protein Engineering/methods , Recombinant Proteins/administration & dosage , Ventricular Dysfunction, Left/prevention & control , Angiogenic Proteins/administration & dosage , Angiogenic Proteins/chemistry , Angiogenic Proteins/genetics , Animals , Delayed-Action Preparations/chemistry , Diffusion , Hepatocyte Growth Factor/analogs & derivatives , Hepatocyte Growth Factor/genetics , Injections , Male , Myocardial Infarction/diagnostic imaging , Myocardial Infarction/pathology , Rats , Rats, Sprague-Dawley , Rats, Wistar , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Shear Strength , Treatment Outcome , Ventricular Dysfunction, Left/diagnostic imaging , Ventricular Dysfunction, Left/pathology , Viscosity
6.
Drug Deliv ; 23(9): 3619-3628, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27562648

ABSTRACT

BACKGROUND: We investigated the therapeutic effectiveness of PEGylated liposomes loaded with angiogenic peptides for treating hindlimb ischemia. METHODS: Rats received a femoral artery occlusion. Red blood cells collected from the animals were labeled with technetium-99m. Limb perfusion gamma imaging was performed. PEGylated liposomes loaded with angiogenic peptides were administered intra-arterially. Technetium-99m red blood cell imaging was repeated 1 week later. The animals were sacrificed the next day. The expression of angiogenic proteins was studied. Later, changes in limb perfusion after intra-arterial infusion versus intra-muscular injection were also compared to determine the therapeutic effectiveness of different administration methods. RESULTS: Femoral artery occlusion dramatically reduced ischemic limb perfusion (by an average of 69%, compared to contralateral limb). This was not different among groups (p > 0.05). Liposomes loaded with angiogenic peptides significantly improved ischemic limb perfusion, compared to controls (210% of baseline, versus 100% of baseline in control; p < 0.05 versus controls). The enhanced ischemic limb perfusion was accompanied by an increased expression of CD 31 (an average of 1.6-fold increase of controls; p < 0.05). The liposomes or peptides treatment alone did not affect ischemic perfusion (liposomes alone: 100% of baseline; peptides alone: 120% of baseline; p > 0.05 versus controls, respectively) or the angiogenic response (1.1-fold of controls in liposomes alone; 1.0-fold of controls in peptides alone; p > 0.05 versus controls, respectively). Intra-muscular injection induced similar liposomal treatment effects on ischemic limb perfusion (230% of baseline) as those by intra-arterial infusion (210% of baseline; p < 0.05 versus intra-muscular). CONCLUSIONS: PEGylated liposomes loaded with angiogenic peptides improved ischemic limb perfusion and promoted angiogenic responses. Liposomal angiogenic treatment via intra-arterial infusion resulted in an equally effective therapeutic efficacy compared to that of intra-muscular injection. These results show the therapeutic potential of our liposomal strategy for treating peripheral limb ischemia.


Subject(s)
Angiogenic Proteins/administration & dosage , Ischemia/drug therapy , Liposomes/administration & dosage , Animals , Cell Line , Drug Administration Routes , Extremities , Femoral Artery/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Male , Perfusion/methods , Polyethylene Glycols/chemistry , Rats , Rats, Sprague-Dawley , Technetium/administration & dosage
7.
Nucl Med Biol ; 43(9): 552-558, 2016 09.
Article in English | MEDLINE | ID: mdl-27395415

ABSTRACT

INTRODUCTION: Although liposomes hold promise for cancer therapy, the effectiveness of treating myocardial ischemia by promoting angiogenesis has yet to be proved. Nanoliposomes loaded with therapeutic agents can effectively target ischemic myocardium via enhanced permeability and retention. Surface polyethylene glycol (PEG) modification can further facilitate effective targeting by prolonging liposomal circulation. This study aimed to determine whether PEGylated nanoliposomes are effective in facilitating targeted drug delivery and treating myocardial ischemia. METHODS: Rats subjected to 30min of myocardial ischemia were given (99m)Tc-hexamethylpropyleneamine oxime- or (99m)Tc-diethylenetriamine pentaacetate-labeled liposomes with mean diameters of ~100nm or ~600nm with or without PEG modifications to determine the extent of myocardial uptake in the different conditions. Therapeutic effectiveness was assessed by studying changes in myocardial perfusion defects with (99m)Tc-tetrofosmin autoradiography and vascular density with immunohistochemistry at 7days post-treatment. RESULTS: The liver and spleen showed the largest capacity for liposome uptake. Uptake by the liver and spleen was more pronounced when the liposomes were larger. Conversely, myocardial liposome uptake was significantly greater when the liposomes were ~100nm rather than ~600nm in diameter. Surface modification with PEG significantly augmented myocardial uptake of ~100nm liposomes. PEG modification did not affect the size dependence. To investigate therapeutic efficacy, hearts subjected to ischemia received PEGylated nanoliposomes encapsulated with angiogenic peptides. Our data demonstrated that PEGylated nanoliposomes loaded with angiogenic peptides improved myocardial perfusion defects and increased vascular density. A 10-fold increase in liposomal concentration did not further benefit myocardial ischemia. CONCLUSIONS: Liposomal angiogenic formulation with size control and PEG modification may be effective treatment strategy for myocardial ischemia. Increasing the concentration of liposomes does not necessarily benefit myocardial ischemia.


Subject(s)
Angiogenic Proteins/administration & dosage , Angiogenic Proteins/pharmacology , Coronary Circulation/drug effects , Myocardial Ischemia/diagnostic imaging , Myocardial Ischemia/physiopathology , Polyethylene Glycols/chemistry , Angiogenic Proteins/therapeutic use , Animals , Capsules , Dose-Response Relationship, Drug , Liposomes , Male , Myocardial Ischemia/drug therapy , Myocardial Ischemia/metabolism , Neovascularization, Physiologic/drug effects , Organophosphorus Compounds/metabolism , Organophosphorus Compounds/pharmacokinetics , Organotechnetium Compounds/metabolism , Organotechnetium Compounds/pharmacokinetics , Radionuclide Imaging , Rats , Rats, Sprague-Dawley , Tissue Distribution
8.
J Control Release ; 217: 191-201, 2015 Nov 10.
Article in English | MEDLINE | ID: mdl-26365781

ABSTRACT

Therapeutic angiogenesis holds great potential for a myriad of tissue engineering and regenerative medicine approaches. While a number of peptides have been identified with pro-angiogenic behaviors, therapeutic efficacy is limited by poor tissue localization and persistence. Therefore, poly(ethylene glycol) hydrogels providing sustained, enzymatically-responsive peptide release were exploited for peptide delivery. Two pro-angiogenic peptide drugs, SPARC113 and SPARC118, from the Secreted Protein Acidic and Rich in Cysteine, were incorporated into hydrogels as crosslinking peptides flanked by matrix metalloproteinase (MMP) degradable substrates. In vitro testing confirmed peptide drug bioactivity requires sustained delivery. Furthermore, peptides retain bioactivity with residual MMP substrates present after hydrogel release. Incorporation into hydrogels achieved enzymatically-responsive bulk degradation, with peptide release in close agreement with hydrogel mass loss and released peptides retaining bioactivity. Interestingly, SPARC113 and SPARC118-releasing hydrogels had significantly different degradation time constants in vitro (1.16 and 8.77×10(-2) h(-1), respectively), despite identical MMP degradable substrates. However, upon subcutaneous implantation, both SPARC113 and SPARC118 hydrogels exhibited similar degradation constants of ~1.45×10(-2) h(-1), and resulted in significant ~1.65-fold increases in angiogenesis in vivo compared to controls. Thus, these hydrogels represent a promising pro-angiogenic approach for applications such as tissue engineering and ischemic tissue disorders.


Subject(s)
Angiogenic Proteins/administration & dosage , Drug Carriers/administration & dosage , Hydrogels/administration & dosage , Neovascularization, Physiologic/drug effects , Oligopeptides/administration & dosage , Osteonectin/chemistry , Angiogenic Proteins/pharmacology , Animals , Bridged Bicyclo Compounds/chemistry , Cells, Cultured , Delayed-Action Preparations/administration & dosage , Delayed-Action Preparations/pharmacology , Drug Carriers/pharmacology , Female , Heptanes/chemistry , Human Umbilical Vein Endothelial Cells , Hydrogels/pharmacology , Matrix Metalloproteinase 2/chemistry , Matrix Metalloproteinase 2/metabolism , Mice, Inbred BALB C , Oligopeptides/pharmacology , Polyethylene Glycols/chemistry
9.
Med Wieku Rozwoj ; 17(1): 31-6, 2013.
Article in English | MEDLINE | ID: mdl-23749693

ABSTRACT

AIM: In this study attention was focused on gene preparations that stimulate angiogenesis in the skin. Angiogenesis was stimulated by gene preparations encoding angiogenic factors introduced into the skin by injection, and applied as ointments. MATERIAL AND METHODS: The appropriate angiogenic formulations containing angiogenic nonviral vectors (pVEGF, pFGF, pSDF, pVIF), or viral vectors (rAAV/VEGF, rAAV/SDF) were prepared for the test. Cholesterol ointment was used as the vehicle for viral and non-viral vectors. The new vessels in the mouse skin were counted according to the criteria suggested by Sidky and Auerbach. RESULTS: Studies indicate that all non-viral (pVEGF, pFGF, pSDF, pVIF) and viral (rAAV/VEGF, rAAV/SDF) vectors strongly stimulate new vessel formation when administered into mouse skin as injections. The impact of angiogenic gene ointments for skin angiogenesis was about 3-4 times weaker than that observed for injection preparations. CONCLUSIONS: Angiogenic injection gene preparations strongly stimulate skin neovascularization. The clinical usefulness of gene ointments should stimulate further laboratory studies in the field of experimental skin gene therapy.


Subject(s)
Angiogenesis Inducing Agents/administration & dosage , Angiogenic Proteins/administration & dosage , Genetic Therapy , Neovascularization, Physiologic/drug effects , Skin/blood supply , Administration, Cutaneous , Animals , Genetic Vectors/administration & dosage , Injections, Intradermal , Mice , Mice, Inbred BALB C , Ointments
10.
Tissue Eng Part A ; 19(3-4): 437-47, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22953721

ABSTRACT

Inflammation and angiogenesis are inevitable in vivo responses to biomaterial implants. Continuous progress has been made in biomaterial design to improve tissue interactions with an implant by either reducing inflammation or promoting angiogenesis. However, it has become increasingly clear that the physiological processes of inflammation and angiogenesis are interconnected through various molecular mechanisms. Hence, there is an unmet need for engineering functional tissues by simultaneous activation of pro-angiogenic and anti-inflammatory responses to biomaterial implants. In this work, the modulus and fibrinogen adsorption of porous scaffolds were tuned to meet the requirements (i.e., ~100 kPa and ~10 nm, respectively), for soft tissue regeneration by employing tyrosine-derived combinatorial polymers with polyethylene glycol crosslinkers. Two types of functional peptides (i.e., pro-angiogenic laminin-derived C16 and anti-inflammatory thymosin ß4-derived Ac-SDKP) were loaded in porous scaffolds through collagen gel embedding so that peptides were released in a controlled fashion, mimicking degradation of the extracellular matrix. The results from (1) in vitro coculture of human umbilical vein endothelial cells and human blood-derived macrophages and (2) in vivo subcutaneous implantation revealed the directly proportional relationship between angiogenic activities (i.e., tubulogenesis and perfusion capacity) and inflammatory activities (i.e., phagocytosis and F4/80 expression) upon treatment with either type of peptide. Interestingly, cotreatment with both types of peptides upregulated the angiogenic responses, while downregulating the inflammatory responses. Also, anti-inflammatory Ac-SDKP peptides reduced production of pro-inflammatory cytokines (i.e., interleukin [IL]-1ß, IL-6, IL-8, and tumor necrosis factor alpha) even when treated in combination with pro-angiogenic C16 peptides. In addition to independent regulation of angiogenesis and inflammation, this study suggests a promising approach to improve soft tissue regeneration (e.g., blood vessel and heart muscle) when inflammatory diseases (e.g., ischemic tissue fibrosis and atherosclerosis) limit the regeneration process.


Subject(s)
Drug Implants/administration & dosage , Guided Tissue Regeneration/instrumentation , Laminin/administration & dosage , Polyethylenes/chemistry , Soft Tissue Infections/therapy , Thymosin/administration & dosage , Tissue Scaffolds , Angiogenic Proteins/administration & dosage , Animals , Anti-Inflammatory Agents/administration & dosage , Equipment Design , Mice , Peptides , Regeneration/drug effects , Soft Tissue Infections/pathology , Treatment Outcome
11.
J Vasc Res ; 50(1): 35-51, 2013.
Article in English | MEDLINE | ID: mdl-23154615

ABSTRACT

Therapeutic angiogenesis is a new revascularization strategy involving the administration of growth factors to induce new vessel formation. The biology and delivery of angiogenic growth factors involved in vessel formation have been extensively studied but success in translating the angiogenic capacity of growth factors into benefits for vascular disease patients is still limited. This could be attributed to issues related to patient selection, growth factor delivery methods or lack of vessel maturation. Comprehensive understanding of the cellular and molecular cross-talk during the different stages of vascular development is needed for the design of efficient therapeutic strategies. The presentation of angiogenic factors either in series or in parallel using a strategy that mimics physiological events, such as concentration and spatio-temporal profiles, is an immediate requirement for functional blood vessel formation. This review provides an overview of the recent delivery strategies of angiogenic factors and discusses targeting neovascular maturation as a promising approach to induce stable and functional vessels for therapeutic angiogenesis.


Subject(s)
Angiogenesis Inducing Agents/administration & dosage , Angiogenic Proteins/administration & dosage , Angiogenic Proteins/genetics , Drug Carriers , Genetic Therapy , Myocardial Ischemia/therapy , Neovascularization, Physiologic , Angiogenic Proteins/biosynthesis , Animals , Delayed-Action Preparations , Humans , Molecular Targeted Therapy , Myocardial Ischemia/genetics , Myocardial Ischemia/metabolism , Myocardial Ischemia/physiopathology , Neovascularization, Physiologic/drug effects , Neovascularization, Physiologic/genetics , Regeneration
12.
Ther Deliv ; 3(6): 693-714, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22838066

ABSTRACT

Therapeutic angiogenesis aims at treating ischemic diseases by generating new blood vessels from existing vasculature. It relies on delivery of exogenous factors to stimulate neovasculature formation. Current strategies using genes, proteins and cells have demonstrated efficacy in animal models. However, clinical translation of any of the three approaches has proved to be challenging for various reasons. Administration of angiogenic factors is generally considered safe, according to accumulated trials, and offers off-the-shelf availability. However, many hurdles must be overcome before therapeutic angiogenesis can become a true human therapy. This article will highlight protein-based therapeutic angiogenesis, concisely review recent progress and examine critical challenges. We will discuss growth factors that have been widely utilized in promoting angiogenesis and compare their targets and functions. Lastly, since bolus injection of free proteins usually result in poor outcomes, we will focus on controlled release of proteins.


Subject(s)
Angiogenesis Inducing Agents/administration & dosage , Angiogenic Proteins/administration & dosage , Drug Delivery Systems , Ischemia/drug therapy , Neovascularization, Physiologic/drug effects , Angiogenesis Inducing Agents/chemistry , Angiogenic Proteins/biosynthesis , Angiogenic Proteins/chemistry , Angiogenic Proteins/genetics , Animals , Chemistry, Pharmaceutical , Delayed-Action Preparations , Drug Carriers , Drug Compounding , Gene Transfer Techniques , Genetic Therapy/methods , Humans , Ischemia/genetics , Ischemia/metabolism , Ischemia/physiopathology , Neovascularization, Physiologic/genetics , Stem Cell Transplantation , Technology, Pharmaceutical/methods
13.
Am J Physiol Heart Circ Physiol ; 303(4): H429-38, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22707563

ABSTRACT

The cytokine midkine (MK) promotes tumor growth mainly by inducing angiogenesis. Here, we identified the source of MK in the vascular system under hypoxic conditions and demonstrated the relevance of MK during ischemia of normal tissue. Hypoxia increased MK protein expression in human polymorphonuclear neutrophils (PMN), monocytes, and human umbilical vein endothelial cells (HUVEC) compared with normoxia. Immunoelectron microscopy showed elevated cell surface expression of MK in PMN and monocytes during hypoxia. However, only HUVEC released significant amounts of soluble MK during hypoxia compared with normoxia (301 ± 81 pg/ml vs. 158 ± 45 pg/ml; P < 0.05). Exogenous MK induced neovascularization in a chorioallantoic membrane (CAM) assay compared with negative control as measured by counting the number of branching points per visual field (1,074 ± 54 vs. 211 ± 70; P < 0.05). In a hind limb ischemia model, the angiogenic response was almost completely absent in MK-deficient mice, whereas control animals showed a profound angiogenic response measured as proliferating endothelial cells per visual field (45 ± 30 vs. 169 ± 34; P < 0.01). These unanticipated results identified endothelial cells as the source of soluble MK in the vascular system during hypoxia and defined MK as a pivotal player of angiogenesis during ischemia in nonmalignant tissue.


Subject(s)
Angiogenic Proteins/metabolism , Chorioallantoic Membrane/blood supply , Cytokines/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Ischemia/metabolism , Muscle, Skeletal/blood supply , Neovascularization, Physiologic , Angiogenic Proteins/administration & dosage , Angiogenic Proteins/deficiency , Angiogenic Proteins/genetics , Animals , Cell Hypoxia , Cell Proliferation , Cells, Cultured , Chick Embryo , Cytokines/administration & dosage , Cytokines/deficiency , Cytokines/genetics , Disease Models, Animal , Hindlimb , Human Umbilical Vein Endothelial Cells/ultrastructure , Humans , Ischemia/genetics , Ischemia/pathology , Ischemia/physiopathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Immunoelectron , Midkine , Monocytes/metabolism , Nerve Growth Factors/administration & dosage , Nerve Growth Factors/metabolism , Neutrophils/metabolism , Time Factors , Up-Regulation , Vascular Endothelial Growth Factor A/administration & dosage
14.
Med Hypotheses ; 79(2): 154-6, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22617757

ABSTRACT

Osteonecrosis of femoral head (ONFH) is a challenging disease. Regardless of underlying causes, the ultimate result in all cases is disruption of femoral head blood supply. Once the disease starts, it is progressive in 80% of cases. Since the majority of the affected individuals are young, every effort should be focused on preserving the patients own femoral head. These years, the role of angiogenic growth factors has been investigated with promising results in animal models of ONFH. Erythropoietin (EPO) is a well known hormone that has been used in treatment of chronic anemia for many years with few side effects. Considering the angiogenic properties of EPO, we hypothesize that local delivery of recombinant human EPO during core decompression will enhance bone regeneration in ONFH. In this way we also can avoid systemic side effects of EPO.


Subject(s)
Bone Regeneration/drug effects , Erythropoietin/administration & dosage , Femur Head Necrosis/drug therapy , Femur Head Necrosis/physiopathology , Models, Biological , Neovascularization, Physiologic/drug effects , Angiogenic Proteins/administration & dosage , Animals , Humans , Injections, Intralesional
15.
J Biomater Appl ; 26(4): 383-417, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21926148

ABSTRACT

Tissue engineering is a newly emerging biomedical technology, which aids and increases the repair and regeneration of deficient and injured tissues. It employs the principles from the fields of materials science, cell biology, transplantation, and engineering in an effort to treat or replace damaged tissues. Tissue engineering and development of complex tissues or organs, such as heart, muscle, kidney, liver, and lung, are still a distant milestone in twenty-first century. Generally, there are four main challenges in tissue engineering which need optimization. These include biomaterials, cell sources, vascularization of engineered tissues, and design of drug delivery systems. Biomaterials and cell sources should be specific for the engineering of each tissue or organ. On the other hand, angiogenesis is required not only for the treatment of a variety of ischemic conditions, but it is also a critical component of virtually all tissue-engineering strategies. Therefore, controlling the dose, location, and duration of releasing angiogenic factors via polymeric delivery systems, in order to ultimately better mimic the stem cell niche through scaffolds, will dictate the utility of a variety of biomaterials in tissue regeneration. This review focuses on the use of polymeric vehicles that are made of synthetic and/or natural biomaterials as scaffolds for three-dimensional cell cultures and for locally delivering the inductive growth factors in various formats to provide a method of controlled, localized delivery for the desired time frame and for vascularized tissue-engineering therapies.


Subject(s)
Tissue Engineering , Angiogenic Proteins/administration & dosage , Animals , Biocompatible Materials , Cell Culture Techniques , Drug Delivery Systems , Humans , Neovascularization, Physiologic , Polymers , Regeneration , Stem Cell Niche , Tissue Engineering/methods , Tissue Engineering/trends , Tissue Scaffolds
16.
Eur Surg Res ; 47(2): 81-9, 2011.
Article in English | MEDLINE | ID: mdl-21720165

ABSTRACT

BACKGROUND: Numerous proangiogenic growth factors have been shown to improve impaired wound healing. This study evaluated the effects of subcutaneous pretreatment with a combination of proangiogenic growth factors on wound closure, mechanical properties, vessel density, and morphology. METHODS: Thirty-six Balb/c mice with streptozotocin-induced diabetes were divided into 3 groups. A mixture of VEGF (35.0 µg), bFGF (2.5 µg), and PDGF (3.5 µg) was administered subcutaneously 3, 5, and 7 days prior to wounding in the first group, whereas the second group received three doses of 3.5 µg PDGF. Wound sizes were assessed daily and the repaired tissues were harvested 7 days after wound closure. RESULTS: Complete closure (≥95% healing of initial wound area) was reached in all proangiogenic pretreated animals by day 17, whereas the PDGF monotherapy group needed up to 20 days for complete closure. By the time of tissue harvesting on day 24, complete closure was not reached in all control animals. Punch biopsy material revealed 1.6-fold higher vessel densities in the proangiogenic combination-pretreated group than in the controls. CONCLUSIONS: Proangiogenic priming revealed several significant effects on diabetic wound healing: faster time to closure, a higher vessel density, and improved functional outcome.


Subject(s)
Angiogenic Proteins/administration & dosage , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/pathology , Wound Healing/drug effects , Animals , Collagen/metabolism , Diabetes Complications/drug therapy , Diabetes Complications/pathology , Diabetes Complications/physiopathology , Diabetes Mellitus, Experimental/physiopathology , Female , Fibroblast Growth Factor 2/administration & dosage , Mice , Mice, Inbred BALB C , Neovascularization, Physiologic/drug effects , Platelet-Derived Growth Factor/administration & dosage , Skin/blood supply , Skin/drug effects , Skin/injuries , Skin/pathology , Skin/physiopathology , Skin Temperature/drug effects , Tensile Strength/drug effects , Vascular Endothelial Growth Factor A/administration & dosage
17.
J Endod ; 37(1): 26-30, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21146071

ABSTRACT

UNLABELLED: Angiogenesis is key to both the development and regeneration of the dentin-pulp complex. OBJECTIVE: We hypothesized that proangiogenic signaling molecules sequestered in dentin matrix can be solubilised to induce angiogenic events. METHODS: Matrix components were extracted from powdered sound human dentin with EDTA and their dose-dependent (0.0001-5 mg/mL) effects examined in endothelial cells in an in vitro angiogenic tube formation assay, proliferation assay, and transcriptional regulation of the VEGF and VEGF-R2 genes. RESULTS: Lower concentrations of dentin matrix components were found to show proangiogenic activity, whereas higher concentrations suppressed angiogenic activity. CONCLUSION: This study highlights that the release of dentin matrix components after dental injury can contribute to the angiogenic events that support pulp regeneration.


Subject(s)
Angiogenic Proteins/physiology , Dentin/physiology , Endothelial Cells/physiology , Extracellular Matrix Proteins/physiology , Neovascularization, Physiologic/physiology , Angiogenic Proteins/administration & dosage , Cells, Cultured , Dose-Response Relationship, Drug , Endothelial Cells/drug effects , Extracellular Matrix Proteins/administration & dosage , Humans , Neovascularization, Physiologic/drug effects , Statistics, Nonparametric
18.
Arterioscler Thromb Vasc Biol ; 31(3): 551-9, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21148423

ABSTRACT

OBJECTIVE: We examined whether plasma levels of angiogenic factors are altered in plasma of patients with peripheral arterial disease (PAD) and whether these factors affect endothelial progenitor cell-induced angiogenesis. METHODS AND RESULTS: Plasma was collected from 184 patients with PAD and 330 age-matched healthy controls. Vascular endothelial growth factor and placental growth factor concentrations did not differ between the groups, whereas we found a linear correlation between PAD disease and thrombospondin (TSP)-1 plasma level. TSP-1 was expressed in newly formed vessels in PAD patients having received local injections of bone marrow mononuclear cells. To analyze the functional role of TSP-1 during neoangiogenesis, we used a Matrigel-plug assay and showed that vascularization of implanted Matrigel-plugs was increased in TSP-1(-/-) mice. Moreover, injections of TSP-1 in C57Bl6/J mice after hindlimb ischemia induced a significant decrease of blood flow recovery. To investigate the effects of TSP-1 on human endothelial colony-forming cell (ECFC) angiogenic potential, recombinant human TSP-1 and a small interfering RNA were used. In vitro, TSP-1 N-terminal part significantly enhanced ECFC adhesion, whereas recombinant human TSP-1 had a negative effect on ECFC angiogenic potential. This effect, mediated by CD47 binding, modulated stromal cell-derived factor 1/CXC chemokine receptor 4 pathway. CONCLUSIONS: TSP-1 is a potential biomarker of PAD and ECFC-induced angiogenesis, suggesting that TSP-1 modulation might improve local tissue ischemia in this setting. ( CLINICAL TRIAL REGISTRATION: NCT00377897.).


Subject(s)
Angiogenic Proteins/blood , Endothelial Cells/metabolism , Ischemia/metabolism , Muscle, Skeletal/blood supply , Neovascularization, Physiologic , Peripheral Arterial Disease/blood , Stem Cells/metabolism , Thrombospondin 1/blood , Angiogenic Proteins/administration & dosage , Angiogenic Proteins/deficiency , Angiogenic Proteins/genetics , Animals , Biomarkers/blood , CD47 Antigen/metabolism , Case-Control Studies , Cell Adhesion , Cell Proliferation , Cells, Cultured , Chemokine CXCL12/metabolism , Disease Models, Animal , Endothelial Cells/transplantation , Hindlimb , Humans , Ischemia/physiopathology , Ischemia/surgery , Mice , Mice, Inbred C57BL , Mice, Knockout , Peripheral Arterial Disease/physiopathology , Peripheral Arterial Disease/surgery , Phenotype , Placenta Growth Factor , Pregnancy Proteins/blood , RNA Interference , Receptors, CXCR4/metabolism , Stem Cell Transplantation , Thrombospondin 1/administration & dosage , Thrombospondin 1/deficiency , Thrombospondin 1/genetics , Treatment Outcome , Vascular Endothelial Growth Factor A/blood
19.
Am J Surg ; 200(5): 655-8, 2010 Nov.
Article in English | MEDLINE | ID: mdl-21056148

ABSTRACT

BACKGROUND: Multilayered alginate microcapsules with a permselective poly-L-ornithine membrane can be used for the dual purpose of encapsulating cells in the inner core and sustained release of angiogenic proteins from the outer layer. The aim of this study was to examine the encapsulation and release of a novel chimeric form of fibroblast growth factor-1 (FGF-1) from the outer layer of alginate microcapsules. METHODS: Heparin-binding growth-associated molecule bound to FGF-1 (HB-GAM/FGF-1) was encapsulated in the outer layer of multilayered alginate microbeads constructed using varying alginate conditions. The encapsulation and release of the chimera was quantified. RESULTS: The outer layer was able to encapsulate and release HB-GAM/FGF-1 for up to 30 days. The outer layer made with 1% alginate of high mannuronic acid content provided the fastest release, while 1.25% high guluronic acid content alginate displayed the longest duration of release. CONCLUSIONS: The outer layer of multilayered alginate microbeads can be used for the encapsulation and long-term release of HB-GAM/FGF-1.


Subject(s)
Alginates/pharmacology , Angiogenic Proteins/administration & dosage , Biocompatible Materials/pharmacology , Carrier Proteins/administration & dosage , Cell Membrane , Cytokines/administration & dosage , Delayed-Action Preparations/pharmacology , Microspheres , Drug Carriers , Glucuronic Acid/pharmacology , Hexuronic Acids/pharmacology , Humans
20.
Macromol Biosci ; 10(11): 1286-301, 2010 Nov 10.
Article in English | MEDLINE | ID: mdl-20857391

ABSTRACT

Functional vascularization is a key requirement for the development and function of most tissues, and most critically cardiac muscle. Rapid and irreversible loss of cardiomyocytes during cardiac infarction directly results from the lack of blood supply. Contractile cardiac grafts, engineered using cardiovascular cells in conjunction with biomaterial scaffolds, are an actively studied method for cardiac repair. In this article, we focus on biomaterial scaffolds designed to mediate the development and maturation of vascular networks, by immobilized growth factors. The interactive effects of multiple vasculogenic factors are discussed in the context of cardiac tissue engineering.


Subject(s)
Coronary Vessels/growth & development , Myocardium/cytology , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Angiogenic Proteins/administration & dosage , Angiogenic Proteins/chemistry , Angiogenic Proteins/pharmacology , Angiogenic Proteins/physiology , Angiopoietin-1/administration & dosage , Angiopoietin-1/chemistry , Angiopoietin-1/pharmacology , Angiopoietin-1/physiology , Animals , Biocompatible Materials/chemistry , Cell Line , Cell Proliferation/drug effects , Collagen/chemistry , Delayed-Action Preparations/chemical synthesis , Delayed-Action Preparations/chemistry , Endothelial Cells/cytology , Endothelial Cells/drug effects , Fibroblast Growth Factors/administration & dosage , Fibroblast Growth Factors/metabolism , Fibroblast Growth Factors/pharmacology , Heparin/chemistry , Heparin/metabolism , Humans , Hydrogels/chemistry , Immobilized Proteins/chemistry , Immobilized Proteins/metabolism , Immobilized Proteins/pharmacology , Mice , Microspheres , Neovascularization, Physiologic/drug effects , Neovascularization, Physiologic/physiology , Vascular Endothelial Growth Factor A/administration & dosage , Vascular Endothelial Growth Factor A/chemistry , Vascular Endothelial Growth Factor A/pharmacology , Vascular Endothelial Growth Factor A/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...