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










Database
Language
Publication year range
1.
Biomaterials ; 45: 56-63, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25662495

ABSTRACT

Hepatocyte growth factor (HGF) has been shown to have anti-fibrotic, pro-angiogenic, and cardioprotective effects; however, it is highly unstable and expensive to manufacture, hindering its clinical translation. Recently, a HGF fragment (HGF-f), an alternative c-MET agonist, was engineered to possess increased stability and recombinant expression yields. In this study, we assessed the potential of HGF-f, delivered in an extracellular matrix (ECM)-derived hydrogel, as a potential treatment for myocardial infarction (MI). HGF-f protected cardiomyocytes from serum-starvation and induced down-regulation of fibrotic markers in whole cardiac cell isolate compared to the untreated control. The ECM hydrogel prolonged release of HGF-f compared to collagen gels, and in vivo delivery of HGF-f from ECM hydrogels mitigated negative left ventricular (LV) remodeling, improved fractional area change (FAC), and increased arteriole density in a rat myocardial infarction model. These results indicate that HGF-f may be a viable alternative to using recombinant HGF, and that an ECM hydrogel can be employed to increase growth factor retention and efficacy.


Subject(s)
Drug Delivery Systems , Hepatocyte Growth Factor/therapeutic use , Hydrogel, Polyethylene Glycol Dimethacrylate/chemistry , Myocardial Infarction/drug therapy , Myocardial Infarction/physiopathology , Protein Engineering , Ventricular Remodeling , Animals , Blood Vessels/drug effects , Blood Vessels/pathology , Cell Size/drug effects , Disease Models, Animal , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Female , Fibrosis/pathology , Heart Function Tests , Humans , Myocardial Infarction/diagnostic imaging , Myocardial Infarction/pathology , Myocytes, Cardiac/pathology , Myocytes, Smooth Muscle/metabolism , Neovascularization, Physiologic/drug effects , Peptide Fragments/pharmacology , Peptide Fragments/therapeutic use , Proto-Oncogene Proteins c-met/metabolism , Rats, Sprague-Dawley , Sus scrofa , Ultrasonography , Ventricular Remodeling/drug effects
2.
Sci Transl Med ; 5(173): 173ra25, 2013 Feb 20.
Article in English | MEDLINE | ID: mdl-23427245

ABSTRACT

New therapies are needed to prevent heart failure after myocardial infarction (MI). As experimental treatment strategies for MI approach translation, safety and efficacy must be established in relevant animal models that mimic the clinical situation. We have developed an injectable hydrogel derived from porcine myocardial extracellular matrix as a scaffold for cardiac repair after MI. We establish the safety and efficacy of this injectable biomaterial in large- and small-animal studies that simulate the clinical setting. Infarcted pigs were treated with percutaneous transendocardial injections of the myocardial matrix hydrogel 2 weeks after MI and evaluated after 3 months. Echocardiography indicated improvement in cardiac function, ventricular volumes, and global wall motion scores. Furthermore, a significantly larger zone of cardiac muscle was found at the endocardium in matrix-injected pigs compared to controls. In rats, we establish the safety of this biomaterial and explore the host response via direct injection into the left ventricular lumen and in an inflammation study, both of which support the biocompatibility of this material. Hemocompatibility studies with human blood indicate that exposure to the material at relevant concentrations does not affect clotting times or platelet activation. This work therefore provides a strong platform to move forward in clinical studies with this cardiac-specific biomaterial that can be delivered by catheter.


Subject(s)
Biocompatible Materials , Extracellular Matrix , Hydrogels/administration & dosage , Myocardial Infarction/therapy , Animals , Swine
3.
Acta Biomater ; 8(10): 3695-703, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22750737

ABSTRACT

Injectable hydrogels derived from the extracellular matrix (ECM) of decellularized tissues have recently emerged as scaffolds for tissue-engineering applications. Here, we introduce the potential for using a decellularized ECM-derived hydrogel for the improved delivery of heparin-binding growth factors. Immobilization of growth factors on a scaffold has been shown to increase their stability and activity. This can be done via chemical crosslinking, covalent bonding, or by incorporating natural or synthetic growth factor-binding domains similar to those found in vivo in sulfated glycosaminoglycans (GAGs). Many decellularized ECM-derived hydrogels retain native sulfated GAGs, and these materials may therefore provide an excellent delivery platform for heparin-binding growth factors. In this study, the sulfated GAG content of an ECM hydrogel derived from decellularized pericardial ECM was confirmed by Fourier transform infrared spectroscopy and its ability to bind basic fibroblast growth factor (bFGF) was established. Delivery in the pericardial matrix hydrogel increased retention of bFGF both in vitro and in vivo in ischemic myocardium compared to delivery in collagen. In a rodent infarct model, intramyocardial injection of bFGF in pericardial matrix enhanced neovascularization by approximately 112% compared to delivery in collagen. Importantly, the newly formed vasculature was anastomosed with existing vasculature. Thus, the sulfated GAG content of the decellularized ECM hydrogel provides a platform for incorporation of heparin-binding growth factors for prolonged retention and delivery.


Subject(s)
Drug Delivery Systems/methods , Extracellular Matrix/metabolism , Fibroblast Growth Factor 2/pharmacology , Heparin/metabolism , Hydrogel, Polyethylene Glycol Dimethacrylate/chemistry , Animals , Arterioles/drug effects , Arterioles/physiology , Elastic Modulus/drug effects , Female , Injections , Microscopy, Electron, Scanning , Neovascularization, Physiologic/drug effects , Pericardium/metabolism , Rats , Rats, Sprague-Dawley , Reproducibility of Results , Rheology/drug effects , Spectroscopy, Fourier Transform Infrared , Sus scrofa
4.
J Am Coll Cardiol ; 59(8): 751-63, 2012 Feb 21.
Article in English | MEDLINE | ID: mdl-22340268

ABSTRACT

OBJECTIVES: This study evaluated the use of an injectable hydrogel derived from ventricular extracellular matrix (ECM) for treating myocardial infarction (MI) and its ability to be delivered percutaneously. BACKGROUND: Injectable materials offer promising alternatives to treat MI. Although most of the examined materials have shown preserved or improved cardiac function in small animal models, none have been specifically designed for the heart, and few have translated to catheter delivery in large animal models. METHODS: We have developed a myocardial-specific hydrogel, derived from decellularized ventricular ECM, which self-assembles when injected in vivo. Female Sprague-Dawley rats underwent ischemia reperfusion followed by injection of the hydrogel or saline 2 weeks later. The implantation response was assessed via histology and immunohistochemistry, and the potential for arrhythmogenesis was examined using programmed electrical stimulation 1 week post-injection. Cardiac function was analyzed with magnetic resonance imaging 1 week pre-injection and 4 weeks post-MI. In a porcine model, we delivered the hydrogel using the NOGA-guided MyoStar catheter (Biologics Delivery Systems, Irwindale, California), and utilized histology to assess retention of the material. RESULTS: We demonstrate that injection of the material in the rat MI model increases endogenous cardiomyocytes in the infarct area and maintains cardiac function without inducing arrhythmias. Furthermore, we demonstrate feasibility of transendocardial catheter injection in a porcine model. CONCLUSIONS: To our knowledge, this is the first in situ gelling material to be delivered via transendocardial injection in a large animal model, a critical step towards the translation of injectable materials for treating MI in humans. Our results warrant further study of this material in a large animal model of MI and suggest this may be a promising new therapy for treating MI.


Subject(s)
Catheterization/methods , Extracellular Matrix/chemistry , Heart Ventricles/metabolism , Hydrogel, Polyethylene Glycol Dimethacrylate/administration & dosage , Myocardial Infarction/drug therapy , Myocytes, Cardiac/pathology , Ventricular Function/drug effects , Animals , Cell Count , Disease Models, Animal , Female , Follow-Up Studies , Heart Ventricles/pathology , Hydrogel, Polyethylene Glycol Dimethacrylate/metabolism , Immunohistochemistry , Injections , Magnetic Resonance Imaging, Cine , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocytes, Cardiac/metabolism , Rats , Rats, Sprague-Dawley , Recovery of Function/drug effects , Swine
5.
Biomaterials ; 30(29): 5409-16, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19608268

ABSTRACT

Myocardial tissue lacks the ability to significantly regenerate itself following a myocardial infarction, thus tissue engineering strategies are required for repair. Several injectable materials have been examined for cardiac tissue engineering; however, none have been designed specifically to mimic the myocardium. The goal of this study was to investigate the in vitro properties and in vivo potential of an injectable myocardial matrix designed to mimic the natural myocardial extracellular environment. Porcine myocardial tissue was decellularized and processed to form a myocardial matrix with the ability to gel in vitro at 37 degrees C and in vivo upon injection into rat myocardium. The resulting myocardial matrix maintained a complex composition, including glycosaminoglycan content, and was able to self-assemble to form a nanofibrous structure. Endothelial cells and smooth muscle cells were shown to migrate towards the myocardial matrix both in vitro and in vivo, with a significant increase in arteriole formation at 11 days post-injection. The matrix was also successfully pushed through a clinically used catheter, demonstrating its potential for minimally invasive therapy. Thus, we have demonstrated the initial feasibility and potential of a naturally derived myocardial matrix as an injectable scaffold for cardiac tissue engineering.


Subject(s)
Biomimetic Materials/administration & dosage , Biomimetic Materials/chemistry , Extracellular Matrix/chemistry , Heart/drug effects , Heart/growth & development , Tissue Engineering/methods , Animals , Biocompatible Materials/chemistry , Injections , Materials Testing , Rats
SELECTION OF CITATIONS
SEARCH DETAIL