Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2015 Dec 10.
Published in final edited form as: J Control Release. 2014 Jul 27;195:110–119. doi: 10.1016/j.jconrel.2014.07.041

Functional Polymers of Gene Delivery for Treatment of Myocardial Infarct

Young-Wook Won 1,2, David A Bull 2, Sung Wan Kim 1,*
PMCID: PMC4252832  NIHMSID: NIHMS618784  PMID: 25076177

Abstract

Ischemic heart disease is rapidly growing as the common cause of death in the world. It is a disease that occurs as a result of coronary artery stenosis and is caused by the lack of oxygen within cardiac muscles due to an imbalance between oxygen supply and demand. The conventional medical therapy is focused on the use of drug eluting stents, coronary-artery bypass graft surgery and anti-thrombosis. Gene therapy provides great opportunities for treatment of cardiovascular disease. In order for gene therapy to be successful, the development of proper gene delivery systems and hypoxia-regulated gene expression vectors are the most important factors. Several non-viral gene transfer methods have been developed to overcome the safety problems of viral transduction. Some of which include plasmids that regulate gene expression that is controlled by environment specific promoters in the transcriptional or the translational level. This review explores polymeric gene carriers that target the myocardium and hypoxia-inducible vectors, which regulate gene expression in response to hypoxia, and their application in animal myocardial infarction models.

Keywords: Myocardial infarct, Gene delivery, Non-viral carrier

1. Introduction

Myocardial infarction (MI) is the leading cause of death in developed nations and one of the most common causes of death in the world. The blockage in coronary arteries by atherosclerosis or thrombus develops ischemic heart disease that includes temporary pain (angina), irregular heart beat (arrhythmia), permanent heart muscle damage (MI), and loss of muscle activity (heart failure) [1]. Cardiac remodeling leading to heart failure is a global and cellular change in ventricular shape and function following chamber dilation, interstitial and perivascular fibrosis. This includes neurohormonal responses, cytokine activation, loss of cardiomyocytes due to necrosis or apoptosis, cardiomyocyte hypertrophy, disruption of extracellular matrix (ECM) and collagen accumulation followed by scar formation [2]. Unfortunately, current pharmacological treatment regimens for myocardial infarction do not reliably limit remodeling of the left ventricle (LV) post-infarction and prevent progression to heart failure [3]. Novel potential treatments, including gene and cell therapies, offer a means to directly treat the pathophysiology underlying the long-term complications of myocardial infarction-loss of cardiomyocytes. The process of remodeling of the left ventricle begins immediately after an acute ischemic insult. The extent of remodeling correlates with the size of the infarct and the decline in cardiac function [4]. Oxidative stress resulting from rapid metabolic changes in the early stages of ischemia plays a crucial role in cardiomyocyte apoptosis and fibrosis of the myocardium [5]. The extent of cardiomyocyte loss in the early stages following an acute MI correlates directly with the subsequent degree of left ventricular remodeling and the decline in cardiac function. This suggests that preventing the loss of cardiomyocytes in the early stages of an acute MI is necessary to achieve long-term efficacy in the treatment of ischemic heart disease.

Since it was first reported in 1972, gene therapy has been a rapidly progressing technology for treating many genetic and acquired diseases including myocardial infarction [6]. The genetic intervention includes (1) overexpression of a target molecule by the introduction of plasmid DNA, (2) a loss-of-function approach by the introduction of RNA interference (RNAi), and (3) correcting deleterious gene mutations/deletions at the genome or primary mRNA level. Neovascularization and the inhibition of apoptosis are considered as good approaches for the sequentially combined gene therapy for ischemic disease. In the early stage of myocardial infarct, reduced oxygen supply and increased reactive oxygen species (ROS) occur in ischemic cardiomyocytes followed by apoptosis. Protecting the cells from apoptosis is the first step, and the second step is to reestablish vasculature through angiogenesis that returns the hypoxic condition back to a normoxic state. DNA, small interfering RNA (siRNA), and micro RNA have been applied to gene therapy. DNA-based gene therapy delivers exogenous plasmid DNA to the cellular nucleus, which encodes a specific gene that enhances the expression of therapeutic proteins. On the other hand, siRNA reduces protein expression by silencing target mRNA in the cellular cytoplasm. However, they must overcome several barriers for successful clinical application such as cell membrane penetration, stability in serum, and safety concerns such as un-controlled gene delivery [7]. To overcome those barriers, DNA and RNA require appropriate delivery vehicles. Various non-viral carriers such as cationic polymers, peptides, liposomes and nanoparticles have been developed and have showed success in the delivery of genes through the cell membrane and into the cell, thus protecting genes from degradation [8].

In 1997, with rationales including a versatile design, no integration into the host chromosome, and non-immunogenic response, research regarding polymeric gene delivery was started [9]. Polymeric carriers are safe for repeated injection, easy to reproduce, and cost-effective, all of which are fundamental considerations in the development of pharmaceutical products [8]. Despite the benefits, they typically show relatively low transfection efficiency and poor therapeutic efficacy compared to virus-mediated gene delivery [10]. Various polymer constructions have been developed to overcome the drawback of polymeric gene carriers. In this review, we describe the use of polymeric gene carriers for treatment of myocardial infarction.

2. Cardiovascular Gene Therapy

A variety of catheter or surgical approaches for in vivo gene transfer into myocardial tissue showed promising results in animal and clinical studies. Specifically, angiogenic gene therapy is of growing interest as an alternative treatment to the conventional protein therapy in the area of ischemic heart diseases. An animal model with chronic ischemic myocardium showed an increase in collateral blood flow and an improvement of cardiac function by the injection of plasmid vascular endothelial growth factor (VEGF) or fibroblast growth factor (FGF) [11, 12]. In human clinical trials, the administration of plasmid VEGF into ischemic myocardium through a left anterior thoracotomy resulted in improved heart responses [13]. Similar to VEGF, administration of FGF into epicardial fat also demonstrated an improvement of angina symptoms and an increase in myocardial blood flow [14]. In addition, anti-apoptotic genes and anti-oxidative genes have been widely used in cardiovascular gene therapy. Despite its great potential as treatment to ischemic heart diseases, the transfection efficiency, stability, safety and controlled expression of the therapeutic genes need to be assured.

3. Polymeric Gene Carriers

A variety of polymer-based gene delivery systems have been developed in the last decade to improve efficacy of therapeutic genes. The polymeric carriers are typically positively charged to bind with the negatively charged the cell membrane that blocks gene transfer into cells [7]. Cationic polymers readily condense negatively charged nucleic acids through electrostatic interaction and have thus been widely used as gene carriers [15-17]. The binding affinity between carriers and nucleic acids may decrease the expression of genes because nucleic acids have to be dissociated from the carrier to move to their target location inside the cells [18]. Designing bioreducible polymers with a proton buffering effect for endosomal escape and rapid dissociation in cytoplasm has solved this problem [19]. Bioreducible polymers, or peptides containing internal disulfide bonds in the main chain, at the side chain or in the cross-linker, have high stabilities in extracellular spaces and are rapidly reduced in the cytosol by high intracellular glutathione (GSH) [20]. Polymer reduction can reduce the cytotoxicity of high molecular weight polycations by converting the polymers back into the smaller constitutive subunits and also allow for the release of nucleic acids into the cytoplasm [21]. The use of bioreducible polymers in gene therapy is increasing due to their potential for enhanced transfection efficiency and cytoplasm-sensitive gene delivery.

4. TerplexDNA

A gene delivery carrier was developed that is derived from stearyl-poly(L-lysine) (stearyl-PLL), low density lipoprotein (LDL) and plasmid DNA. TerplexDNA is generated through electrostatic and hydrophobic interactions between LDL, stearyl-PLL, and DNA [22, 23]. The PLL component condenses DNA through the interaction of the epsilon-amino group of the lysine with the negatively charged phosphate backbone of DNA [22, 23]. The stearyl groups participate in hydrophobic interactions with the core of the LDL molecule allowing integration into the molecule itself [24]. Incorporation of LDL into the polymer gene carrier enhances gene delivery through augmentation of the LDL receptor-mediated endocytosis pathway. LDL receptors exist on the surface of many cells including artery endothelial cells, myocytes, and hepatocytes [25, 26].

Compared with Lipofectamine, the TerplexDNA system showed high transfection efficiency without cytotoxicity in a human hepatocyte line (HepG2), murine smooth muscle (A7R5) cell lines [22, 23], and bovine aorta primary cell cultures, both vascular smooth muscle cells and endothelial cells [24]. Pharmacokinetic and biodistribution studies revealed that TerplexDNA improved circulation time and prolonged whole body retention. TeplexDNA was less toxic than other cationic polymers because the mechanism of internalization is receptor-mediated endocytosis [23, 27]. The main advantage of TerplexDNA might be due in part to the prolonged half-life, which provides the benefit of an increased chance of interaction with the LDL receptor. Another advantage might be the possibility of repeated administration because TerplexDNA is non-immunologic [23, 27]. The TerplexDNA system may therefore have applications in the treatment of heart disease in a clinical setting.

4.1 Gene Delivery into Myocardium

The TerplexDNA system would be beneficial for delivering DNA to the myocardium because the LDL receptors exist on the surface of myocytes [28]. The transfection efficiency of TerplexDNA in rabbit myocardium was 20-100-fold higher than that of naked DNA. This system exhibited more widespread and uniform gene expression near the injection area. In a rat myocardial infarction model, gene transfection was significantly improved without toxicity when compared to naked DNA. TerplexDNA system was developed as an efficient gene carrier that has potential in future clinical applications for the treatment of cardiovascular diseases.

4.2 Gene Delivery into Primary Artery Wall Cells

Receptor-mediated endocytosis was found to be the main mechanism of TerplexDNA internalization into cells [22, 23]. Along with the fact that the cells of the artery wall also express LDL receptors on their cell surface [25], it was hypothesized that TerplexDNA system specifically delivers genes into these cells. This TerplexDNA system was evaluated with these cells for transfecting reporter genes (β-galactosidase and luciferase) or VEGF gene. TerplexDNA system specifically delivered reporter genes and VEGF gene into the cells of the bovine aortic artery wall by receptor mediated endocytosis [24]. The gene transfection by TerplexDNA system was significantly inhibited in the presence of free LDL. This proves that TerplexDNA system is a promising tool for artery wall gene transfer.

4.3 VEGF Gene Delivery in Animal Model

The mechanism of left ventricular dysfunction after myocardial infarction is multifactorial and not completely understood [29]. It was reported that the essential role of myocytes that secrete VEGF was to maintain the function of the left ventricle [30]. This finding suggests the autocrine and paracrine role of VEGF in the maintenance of normal function of the myocardium and the left ventricular extracellular matrix, which can be noted as an important factor to prevent the progress of myocardial infarction. The Protective effects of VEGF on the left ventricular function were proved in an ex vivo model of acute ischemia [31]. Positive therapeutic efficacy was observed from an animal model that was administered TerplexDNA system delivering plasmid VEGF [32]. The improvement in left ventricular function in the VEGF-treated animals was accompanied by an inhibition of the usual increase in left ventricular systolic and diastolic area as seen commonly following a myocardial infarction.

5. Water Soluble Lipopolymer (WSLP)

WSLP was synthesized by combining the cationic head group of branched PEI (bPEI 1.8 KDa) with a hydrophobic lipid anchor, cholesterol chloroformate. It was reported as an efficient gene delivery carrier [33-35]. High molecular weight branched PEI (bPEI 25 kDa) has been known as a cationic polymer that has shown high transfection efficiency due to the proton buffering effect. However, bPEI25 kDa is highly toxic to cells [36]. Low molecular weight bPEI such as bPEI1800 is less cytotoxic, but shows low efficiency in gene transfection. WSLP has many advantages over PEIs. First, WSLP contains the PEI moiety, which has the proton buffering effect. This buffering effect facilitates endosome escape, resulting in high transfection efficiency. Second, WSLP is less toxic to cells because it contains bPEI1800. Third, the cholesterol moiety in WSLP enhances transfection efficiency. Cholesterol is taken up by the cells through receptor-mediated endocytosis or transfer from the lipoprotein to the exoplasmic leaflet of the plasma membrane bilayer [37]. Fourth, WSLP does not require a co-lipid, unlike other cholesterol-based cationic lipids. Since WSLP can escape the endosome, other co-lipids that destabilize the endosomal membrane are not necessary [33, 34, 38].

5.1 Gene Delivery into Myocardium

WSLP was evaluated as a gene carrier to smooth muscle cells in vitro and to the myocardium in vivo [34]. The transfection efficiency of WSLP was higher than that of PEI1800, lipofectamine, or SuperFect to A7R5 smooth muscle cells. In addition, WSLP showed negligible cytotoxicity to A7R5 cells. The uptake mechanism studies suggested that WSLP internalized into cells through a cholesterol-mediated uptake mechanism [34]. Moreover, the injection of WSLP/pDNA to rabbit myocardium also showed that WSLP had higher transfection efficiency than PEI1800.

5.2 VEGF Gene Delivery in Animal Model

The construction of an effective plasmid DNA that encodes a specific therapeutic gene is important for successful gene therapy. VEGF has been known to be the most effective therapeutic protein for the neovascularization [39]. However, Springer et al. proved that exogenously delivered VEGF could exert a physiological effect in normal, nonischemic tissues [40]. This suggested that VEGF expression must be carefully modulated. In addition, unregulated continuous expression of VEGF is associated with the formation of endothelial cell-derived intramural vascular tumors [41].

Erythropoietin (Epo) enhancer was previously reported to enhance the activity of the promoter under hypoxia conditions [42-44]. Those studies showed that the Epo enhancer induced reporter gene expression in various cell lines under hypoxia, suggesting that the Epo enhancer could regulate gene expression. Therefore, the Epo enhancer is a good gene regulator to express VEGF in response to hypoxia. A hypoxia-inducible plasmid expressing VEGF was constructed by adding the Epo enhancer in the upstream of the VEGF coding region [35]. The efficiency of the pEpo-SV-VEGF/WSLP complex was evaluated in vitro and in vivo. The promoter has a hypoxia-responsive element that binds to hypoxia-inducible factor-1 (HIF-1) and activates the transcription of the gene under hypoxia conditions [45-47]. After transcription, the VEGF mRNA is stabilized by cooperation of multiple RNA elements such as coding regions and 5′- and 3′-untranslated regions (UTRs), resulting in an increase of the translation rate [48]. Several hypoxia-regulated genes including Epo have hypoxia-responsive elements in their promoters and enhancers. By employing the Epo enhancer for the regulation of the VEGF gene, we mimicked the natural regulation of VEGF expression. Hypoxia-regulated gene expression in the mouse ischemic heart model was reported using a hypoxiaresponsive element by adeno-associated virus-mediated gene transfer [49]. In addition, the Epo-enhancer-mediated induction of the VEGF gene was proven in a rabbit ischemic heart model. These results suggested that the hypoxia-inducible VEGF gene delivery system in combination with WSLP is effective and safe for the treatment of ischemic heart disease.

Springer et al. proved that exogenously delivered VEGF could exert an unknown effect in normal and non-ischemic tissues [40]. In addition, unregulated continuous expression of VEGF is associated with formation of endothelial cell–derived intramural vascular tumors [41]. These findings suggest that VEGF expression must be regulated. It has been shown that Epo enhancer-SV40 promoter induced gene expression in vitro under hypoxia and in rabbit ischemic myocardium in vivo [35]. In addition, Su et al. proved the over-expression of the hypoxia-responsive element-induced VEGF in ischemic myocardium by using an adeno-associated virus [49]. Hypoxia-specific regulation of the VEGF expression system may be useful for safer VEGF gene therapy by minimizing unwanted side effects.

6. Ischemic Myocardium-Specific Gene Regulation Systems

6.1 RTP801 Promoter

The RTP801 promoter is an effective regulatory system in response to hypoxia. Shoshani et al. reported that RTP801 transcription was rapidly and sharply increased both in vitro and in vivo under hypoxic condition [50]. This inducible expression of RTP801 is mediated by transcriptional activation. In addition, it was suggested that the induction of the RTP801 promoter was mediated by HIF-1, which is a transcription factor that mediates hypoxia induction of a number of genes [51, 52]. Binding of HIF-1 to the consensus domain of the genes results in the transcriptional induction of the gene promoters [43, 53]. HIF-1–mediated induction of gene transcription is a widespread oxygen-sensing mechanism in various types of cells [44].

The RTP801 promoter was analyzed and identified a cis-regulatory element that was responsible for the hypoxia induction of the promoter [54]. A potential Sp1 element in the RTP801 promoter was involved in hypoxia induction. It was shown that the RTP801 promoter was induced under hypoxia conditions in various types of cells. A VEGF plasmid including the RTP801 promoter controlled VEGF expression under hypoxia. This promoter could minimize the side effect of the nonspecific VEGF expression and induce VEGF expression specifically in ischemic tissue. The results suggested that a hypoxia-inducible VEGF plasmid, pRTP801-VEGF, might be a useful angiogenic gene for the treatment of ischemic diseases.

In another study, delivery of RTP-VEGF plasmid using a novel reducible disulfide poly(amido ethylenediamine) (SS-PAED) polymer carrier was studied in vitro and in vivo [55]. In vitro transfection into rat cardiomyocytes (H9C2) showed that SS-PAED increased the gene expression by 16-fold compared to bPEI control. SS-PAED mediated delivery of RTP-VEGF plasmid produced significantly higher levels of VEGF expression (up to 76 fold) under hypoxic conditions compared to normoxic conditions in both H9C2 and rat aortic smooth muscle cells (A7R5). Using SS-PAED, delivery of RTP-VEGF was investigated in a rabbit myocardial infarct model. Results showed up to 4-fold increase in VEGF protein expression in the region of the infarct compared to injections of SS-PAED/RTP-Luc.

Although unregulated, constitutively expressed VEGF gene therapy reduced myocardial infarct size, ischemia-inducible VEGF gene therapy demonstrated even greater efficacy. The mechanism for the improved efficacy seen with the ischemia-inducible VEGF gene therapy is as follows: (1) decreased apoptosis and (2) increased angiogenesis, compared with unregulated, constitutively expressed VEGF gene therapy following myocardial infarction. It is likely that both mechanisms contribute to the significant decrease in myocardial infarct size. In addition to these two mechanisms, VEGF gene therapy has previously been shown to (1) promote arteriogenesis and (2) have a mitogenic effect on adult cardiomyocytes in large animal models of myocardial ischemia and infarction [56-58]. Both of these previously described mechanisms most likely also contribute to the significant decrease in myocardial infarct size.

6.2 Epo Enhancer

The activity of the Epo enhancer-SV40 promoter system was further enhanced without significant decrease in its specificity by co-transfection of HIF-1-alpha gene [59]. Co-transfection of pSV-HIF1 alpha and pEpo-SV-Luc increased the promoter activity of the Epo enhancer-SV40 promoter system, showing at least a 3-fold higher gene expression under hypoxia as compared with the pEpo-SV-Luc single-plasmid transfection. Furthermore, co-transfection showed significant hypoxia specificity. Also, co-transfection of pEpo-SV-VEGF with pSV-HIF-1-alpha showed enhanced VEGF expression without loss of hypoxia specificity when compared with pEpo-SV-VEGF single-plasmid transfection. Furthermore, pSV-HIF-1-alpha induced the endogenous hypoxia-responsive genes such as angiopoietin-1, which would be beneficial for therapeutic angiogenesis. Therefore, with hypoxia specificity and higher gene expression, co-transfection of pSV-HIF-1-alpha and pEpo-SV-VEGF may be useful for ischemia targeting gene therapy.

6.3 Post-translational Gene Regulation System

Hypoxia-responsible gene expression can be primarily regulated in three ways: 1) transcriptional regulation; 2) post-transcriptional regulation, and 3) post-translational regulation [60]. HIF-1 is accepted as the most important element in transcriptional regulation. In post-transcriptional regulation, VEGF mRNA is stabilized under hypoxic conditions by the cooperation of the 5′- and 3′-untranslated regions (UTRs) and the coding region. The best-studied strategy to control gene expression under hypoxic conditions in post-translational regulation is the fusion of the oxygen-dependent degradation (ODD) domain and the therapeutic gene in order to stabilize the protein produced [61, 62]. The use of the ODD domain constitutes a powerful tool to increase the resident time of proteins, which typically have a short half-life. VEGF in particular needs to be stabilized following expression because its half-life is very short [63-65]. Unlike the RTP801, the pβ-SP-ODD-VEGF composed of the signal peptide, the ODD domain, and the furin-cleavage site regulates VEGF secretion in response to hypoxic conditions at the post-translational level [66]. The ODD domain stabilizes VEGF in hypoxic cells, while the furin site is cleaved by furin enzyme in the Golgi apparatus, leading to the production of wild-type VEGF. The SP domain then facilitates the exogenous secretion of this wild-type VEGF, which is known as the most potent form of VEGF [67, 68].

While the presence of the ODD domain can play a positive role in post-translational regulation, its large molecular weight may cause an abnormal folding or a decrease in secretion of the therapeutic protein, thereby diminishing the efficacy of the secreted therapeutic protein [60, 69]. These potential shortcomings may be overcome by the use of a short ODD domain composed of 18 core amino acids [69]. However, this shorter ODD domain may interfere with the interaction between VEGF and its receptors. Enzymatic degradation between the ODD domain and VEGF helps to ensure that the wild-type form of VEGF is secreted following processing in the Golgi network [70].

Furin, a member of the subtilisin-like proprotein convertase family that exists in the Trans Golgi Network (TGN), processes latent precursor proteins into their biologically active products in the secretory pathway [71]. Furin recognizes a conserved polybasic R-X-R/K-R site and cleaves that site downstream of the target sequence [72, 73]. R-G-R-R, a furin-recognition site, was inserted between the ODD domain and the VEGF-coding region to enhance the secretion of wild type VEGF [74]. The decrease in VEGF secretion seen with the increase in the concentration of the furin inhibitor suggests that VEGF secretion is accelerated by the cleavage of the signal peptide (SP) and the ODD domains. The finding that the furin inhibitor hinders VEGF secretion and increases the intra-cellular concentration of VEGF also confirms that the furin-recognition site is a powerful tool to enhance the secretion of wild-type VEGF.

A plasmid containing the SP domain, the ODD domain, and the furin-cleavage site is a promising construct for enhancing VEGF secretion and the activity of VEGF under hypoxic conditions [66]. The SP domain directs VEGF into the secretory pathway while the ODD domain stabilizes the expression of VEGF. The furin-recognition site provides a specific region to separate VEGF from the SP and ODD domains, resulting in more efficient secretion of wild-type VEGF. This pβ-SP-ODD-VEGF plasmid was more efficient to produce wild-type VEGF than the pRPT-VEGF plasmid, demonstrating that this plasmid construct is superior to other hypoxia-inducible VEGF plasmids. These findings suggest that pβ-SP-ODD-VEGF may be a promising gene construct for the treatment of a variety of clinically important ischemic disease states.

The protective effects of VEGF gene therapy in the setting of acute myocardial ischemia are due not only to the induction of angiogenesis but also to the prevention of apoptosis of the cardiomyocytes. The extent of the inhibition of apoptosis is directly correlated with the amount of VEGF released [75, 76]. The significant reduction in the number of apoptotic cardiomyocytes and the size of the myocardial infarcts in the dendrimer type-arginine-grafted bioreducible poly(CBA-DAH) (ABP) PAM-ABP/pβ-SP-ODD-VEGF treated hearts demonstrate that the pβ-SP-ODD-VEGF is more efficacious at preventing left ventricular remodeling and preserving left ventricular function than the RTP-VEGF. This is consistent with previously published VEGF secretion data [66] showing more efficient secretion of wild-type VEGF using the pβ-SP-ODD-VEGF plasmid compared to the RTP-VEGF plasmid.

The difference in efficacy between the RTP-VEGF and the pβ-SP-ODD-VEGF is, at least in part, due to where the hypoxia-responsive regulation of gene expression occurs. The RTP801 promoter includes HIF-1 binding sites and stimulating protein-1 (Sp1) that is up regulated under hypoxic conditions. Formation of a multi-protein complex of Sp1 with HIF-1 and Smad3, used as a co-activator and adaptor protein, results in an Sp1-Smad3-HIF-1 complex on the RTP801 promoter that works to enhance gene expression under hypoxic conditions [60, 77]. This hypoxia-responsive regulation of VEGF expression and secretion is the mechanism underlying the differences in the therapeutic effects of the pβ-SP-ODD-VEGF and the RTP-VEGF observed in previous studies [78]. It has been demonstrated that hypoxia-responsive VEGF gene therapy improves left ventricular function and prevents left ventricular remodeling following acute myocardial ischemia by inhibiting apoptosis of cardiomyocytes and promoting angiogenesis. When comparing two different hypoxia-responsive regulation systems, i.e. transcriptional versus post-translational regulation, pβ-SP-ODD-VEGF, the post-translationally regulated hypoxia-responsive plasmid was more efficacious than RTP-VEGF, a transcriptionally regulated hypoxia-inducible system, at improving the effects of ischemia/reperfusion injury in a rat model. The PAM-ABP/pβ-SP-ODD-VEGF shows promise as a potential novel therapy for the treatment of myocardial ischemia and infarction.

6.4 Combined Gene Delivery

Single gene therapy has failed to prevent the lethal arrhythmias, acute cardiogenic shock, and chronic end-stage heart failure, which are the potentially fatal complications of ischemic heart disease [79]. Sequentially combined gene therapy is a promising treatment for ischemic diseases because the single gene therapies are effective for either preventing apoptosis of ischemic cells or inducing angiogenesis. Gene therapy for ischemic diseases requires the induction of neovascularization and the inhibition of apoptosis. In addition, successful ischemic disease gene therapy via the above approaches needs sufficient genetic interventions based on a precise basic understanding of the mechanisms of heart failure.

A hypoxia-inducible plasmid was constructed for the dual expression of heme oxygenase-1 (HO-1; knock-in, anti-oxidation) and the Src homology domain-2 containing tyrosine phosphatase-1 (SHP-1) microRNA (miSHP-1; knockdown, anti-apoptosis). In addition to the anti-oxidative and the anti-apoptotic effects of HO-1 and mi SHP-1, several studies have reported that the overexpression of HO-1 and the silencing of SHP-1 accelerate angiogenesis in ischemic myocardium [80-82]. HO-1, a stress-inducible antioxidant enzyme, exerts potent cardioprotective effects through its anti-inflammatory, anti-apoptotic, and anti-oxidant activity in ischemic tissue [83]. HO-1 gene therapy may protect the heart from ischemia/reperfusion injury by suppressing the early inflammatory response and inhibiting cardiomyocyte apoptosis [84].

SHP-1, a key molecular mediator of apoptosis, negatively regulates anti-apoptotic signaling pathways, including extracellular signal-regulated kinase (ERK1/2) and BCL-2. SHP-1 binding to death receptors such as TNFR-1 and FAS-R promotes apoptosis through the regulation of de-phosphorylation in signal transduction pathways [85, 86]. Decreased cardiomyocyte apoptosis and increased cardio-protection through Akt activation by the inhibition of the SHP-1 gene suggest that a therapeutic strategy designed to inhibit the expression of SHP-1 by miRNA would be effective in IHD [87, 88]. Although miSHP-1 can reduce apoptosis and protect cells in ischemic tissue by inhibiting SHP-1 gene expression, induction of elevated levels of RNAi may be toxic to cells due to the interference with intrinsic cellular RNAi processes. Unlike siRNA, miRNA production can be regulated by promoters/enhancers, which transcribe genes in response to specific intracellular environments or signals, ensuring that RNAi activity occurs in specific tissues or cell conditions [89].

The hypoxia-inducible plasmid for the dual expression of HO-1 and miSHP-1 demonstrated higher levels of HO-1 expression and mature miSHP-1 production in hypoxic cells compared to normoxic cells [90]. In addition, both HO-1 and miSHP-1 can synergistically enhance the secretion of VEGF. Several studies have reported that both HO-1 overexpression and SHP-1 silencing accelerate angiogenesis in ischemic areas [80-82]. The combined gene delivery demonstrated in these studies is promising for ischemic diseases in terms of not only synergistic efficacy but also sequential activities that prevent cardiomyocyte apoptosis and induce angiogenesis [91] because antiapoptotic genes typically protect cells in ischemic tissues for up to 2 weeks, and neovascularization requires more than 1 week upon VEGF secretion [92]. This sequentially and synergistically combined gene therapy provides the double effects of cardiomyocyte protection in the early stage of ischemia and vascular regeneration in the late stage.

7. Targeted Gene Delivery into Myocardium

7.1 Artery Wall-Targeted Gene Delivery

Apolipoprotein B-100 (apo B-100), a major protein component of LDL, contains many receptor-binding domains such as LDL receptor-binding domain, artery wall cell-binding domain, and heparin-binding domain. Shih et al. demonstrated that a synthetic peptide containing 1000–1016 amino acid residues of apo B-100 (Arg-Ala-Leu-Val-Asp-Thr-Leu-Lys-Phe-Val-Thr-Gln-Ala-Glu-Gly-Ala-Lys) was the arterial wall-binding domain of apo B-100 [26]. The focal accumulation of 125I-labled apo B-based synthetic peptide at the healing edges of regenerating endothelial islands in balloon-cather deendothelialized rabbit aorta suggested that this arterial wall-binding peptide could mediate accumulation of LDLs in arterial lesions. A synthetic peptide was selected based on apo B-100 and added cysteine at the N-terminus of the peptide to facilitate conjugation to PEG-g-PLL [93]. An artery wall-targeted gene delivery system based on PEG-g-PLL was synthesized by introducing the artery wall binding peptide (AWBP) to the end of PEG-g-PLL. The AWBP provided a means of targeting to the arterial wall cells and was shown to locate at the target site for a sufficient amount of time to treat the genetic problem. Luciferase gene transfer using AWBP-PEG-PLL confirmed that the targeted gene delivery to bovine aorta wall cells was mediated by specific artery wall cell receptor-mediated endocytosis.

7.2 Prostaglandin E2 (PGE2) targeting

Specific ligand-modified gene delivery systems are promising strategies to successfully control gene expression in target cells. The specific ligands bind to cell surface receptors and increase cellular uptake of therapeutic genes via target receptor-mediated endocytosis [94]. There have been many cell-selective siRNA delivery systems constructed using specific ligands for cancer or liver cells [95, 96]. Cardiovascular targets for siRNA therapy, however, have no specific vector system, except physical targeting methods. PGE2 is involved in numerous physiological mechanisms including the contraction and relaxation of smooth muscle, control of blood pressure, and modulation of inflammation [97]. Its diverse physiologic effects are exerted via four distinct PGE2 receptors (EP1-4) located on the cell surface [98], and PGE2 receptors (EPs) participate in agonist-induced internalization upon PGE2 stimulation [99]. Although the expression levels of EPs vary between different species, EP4 has been reported to be highly expressed in the hearts of several species, including humans [100]. Accordingly, it was hypothesized that the use of PGE2 as a specific ligand for cardiovascular-targeted siRNA delivery would increase the efficiency of siRNA transfer to cardiomyocytes.

Linear and branched types of reducible cationic copolymers (disulfide-containing poly(amido amine)s, SS-PAAs), synthesized by Michael-type addition between various amine-containing monomers and cystamine bis-acrylamide, have been reported as effective gene carriers [55, 101-103]. A linear SS-PAA, synthesized by copolymerization between 1,6-diaminohexane and cystamine bis-acrylamide (cystamine bisacrylamide-diamino hexane (poly(DAH/CBA)), served as a powerful vector to carry Fas siRNA into rat cardiomyocytes [104]. PGE2 was conjugated to Fas siRNA molecules, and the synthesized PGE2–Fas siRNA contributed to cardiomyocyte targeting via PGE2 receptor-mediated intracellular delivery. Cardiomyocyte apoptosis is the leading cause of heart failure after myocardial infarction. It has been reported that many apoptosis-related genes including Fas/Fas ligand are overexpressed in apoptotic cardiomyocytes. For successful in vivo application of siRNA therapeutics, it is necessary to develop a cardiomyocyte-targeted siRNA delivery system with high transfection efficiency. These results suggest that cardiomyocyte-targeted Fas siRNA delivery using the PGE2–Fas siRNA/poly(DAH/CBA) polyplex is a promising approach to inhibit apoptosis in the treatment of cardiovascular disease.

7.3 Primary Cardiomyocyte (PCM) Targeting

PCM is an isolated phage that displays a 20 amino acid peptide (WLSEAGPVVTVRALRGTGSW), which binds to primary cardiomyocytes 180 times more avidly than control phages [105, 106]. The PCM peptide sequence was used as a specific cardiomyocyte-targeting ligand for the delivery of genes and the potential treatment of cardiovascular disease. The Fas has been identified as an inducer of cardiomyocyte apoptosis, and many studies using Fas siRNA have demonstrated that inhibition of Fas, in turn hinders cardiomyocyte apoptosis without immune stimulation [107-109]. The ability of a synthesized PCM-modified bioreducible polymer containing Fas siRNA to down-regulate Fas gene expression and inhibit cardiomyocyte apoptosis has been described previously [110]. The study was designed to deliver siRNA effectively to a specific target, cardiomyocytes, with a high transfection efficiency, by modifying siRNA with PCM or cell-penetrating (Tat) peptides [111]. These peptide-conjugated siRNAs did not initially produce dense nanoparticles with CBA-DAH, but the addition of plasmid DNA resulted in a more stable and compact polyplex formation. The resulting compact C-siRNA-pDNA/CD polyplexes promoted high levels of cellular uptake and effective gene silencing in cardiomyocytes without significant immunogenicity. In addition, the combined siRNA polyplexes, that is, C-siRNA with PCM-siRNA and Tat-siRNA, were delivered to the heart at significantly higher levels compared to the unmodified siRNA following systemic administration. These findings indicate that siRNA-pDNA/CD complex may be potentially useful therapeutic tool for cardiomyocyte specific gene therapy.

7.4 Ischemic Myocardium Targeting

Systemic administration of polyplexes targeted to the ischemic myocardium affords significant advantages over local injection. First, targeted polyplexes can be administered immediately following reperfusion via iv injection and do not require a separate procedure with accompanying risks to deliver the polyplex locally. Second, systemic administration allows for easy follow-on dosing to maximize the gene therapy benefit. A primary cardiomyocyte-targeted polymeric gene carrier was developed that enhanced gene transfection in cardiomyocytes using a homing peptide [110]. However, since it may direct the gene to alternative heart tissues as well as the intended ischemic region when administered systemically, future challenges still remain [105]. Recently, a peptide sequence that has high specificity to ischemic myocardium was identified by in vivo phage display in the ischemia/reperfusion (I/R) rat model [112]. In vivo phage display for high-throughput screening, referred to as in vivo biopanning, allows for the screening of a specific peptide that homes organs or tissues in a living animal [113]. This technique is useful for the development of targeted therapeutics, imaging agents, and diagnostic markers in various diseases [114]. Nevertheless, only a few studies have reported targeted delivery of therapeutics by using diseased tissue-targeted peptides, and their main objectives were targeting to tumors or blood vasculature. It has been demonstrated that the ischemic myocardium-targeted peptide (IMTP) could direct specific genes to the ischemic heart and increase the accumulation and transfection of said genes in the ischemic myocardium.

CBA-DAH was chosen as a backbone for the modification with IMTP and d-9-arginine (9R). Since 9R has been known as the most effective peptide for protein transduction as well as gene transfection, the attachment of 9R to CBA-DAH would increase the transfection efficiency and decrease the amount of polymer required for sufficient transfection [115-117]. Although various polymeric gene carriers have shown evidence of a great potency for cardiovascular medicine, gene therapy for cardiovascular disease still has challenges due to the difficulty in ischemic heart targeting [118]. The targeting effect of IMTP-CD-9R/DNA polyplex and increase in gene expression in the LV in an I/R rat model after a systemic injection has been clearly demonstrated [119]. An ischemic myocardium-targeted gene delivery system was developed by conjugation of IMTP to CBA-DAH polymer, which reduced the required amount of polymer for transfection by the attachement of 9R. Conclusively, IMTP-CD-9R is capable of targeting to the ischemic myocardium and enhances gene expression in the LV in an I/R rat model upon an intravenous injection.

8. Genetically Engineered Cell Delivery

A novel approach to cardiac repair is to combine cellular transplantation and angiogenic gene therapy. The gene carriers that can transduce angiogenic genes into primary myoblasts, however, have not been optimized. While viral vectors, including adenovirus, adeno-associated virus, and retrovirus, have high gene transfection efficiency, they have limited clinical application due to their inherent potential for immunogenicity, tumorigenicity, induction of an inflammatory response, and integration into the host genome [120-122]. Furthermore, long-term over-expression of VEGF via viral vectors has been observed to lead to hemangioma instead of functional vessels in animal models [41, 123]. These findings exclude viruses as clinically viable vectors to transduce angiogenic genes into primary myoblasts. It is therefore necessary to develop a safer and more efficient non-viral gene carrier that can circumvent the limitations of viral vectors.

The in vitro gene expression efficiency and therapeutic effectiveness of polymer mediated transfection of primary myoblasts has been previously assessed [124]. Autologous primary myoblast transplantation may improve the function of infarcted myocardium via myogenesis. In addition, primary myoblasts can carry exogenous angiogenic genes that encode angiogenic factors to promote therapeutic angiogenesis. Two biodegradable poly(disulfide amine)s, CBA-DAH and CBA-DAH-arginine (CBA-DAH-R), were synthesized as polymer carriers for gene delivery. The in vitro time-course and co-culture experiments verified that polymer engineered primary myoblasts have the ability to stimulate endothelial proliferation. These findings confirmed that poly (disulfide amine)s are the safe and feasible polymeric gene carriers to transfect VEGF 165 into primary myoblasts. Polymer engineered primary myoblasts have potential for therapeutic application in the treatment of ischemic heart diseases.

Exogenous primary myoblast transplantation into ischemic myocardium offers many potential benefits for the treatment of myocardial infarction, including improvement and restoration of cardiac function. The improvement in cardiac function is thought to be due to the enhancement of the contractile properties of endogenous cardiomyocytes and the differentiation and proliferation of implanted myoblasts into functional myocardium within ischemic hearts. Recent studies have demonstrated the feasibility of primary myoblast transplantation for the treatment of ischemic heart disease [125, 126]. In addition, it has been previously reported that VEGF gene therapy, particularly ischemia-inducible VEGF gene therapy, significantly improves myocardial function and reduces left ventricular infarct size following acute myocardial infarction [32, 127]. Excessive VEGF expression, however, has been associated with heart failure and death, due to vascular tumor formation instead of the formation of functioning vessels [128]. Previous work has laid the foundation for current studies demonstrating the functional benefit of this approach for treating acute myocardial infarction in an in vivo rat model [124]. Treatment with poly (CBA-DAH)/VEGF-transfected myoblasts preserved cardiac wall thickness, restored left ventricular function, induced neovascularization and reduced cardiac apoptosis more effectively than untransfected myoblasts. This work demonstrates that combining cellular implantation therapies with gene therapy can potentially increase the efficacy of surgical implantation of cells and produce better patient outcomes.

9. Minicircle DNA encoding VEGF

The application of plasmid DNA-based gene therapy is limited by its inefficient transgene expression. Minicircle DNA was evaluated for efficient VEGF expression in skeletal muscle cells [129]. The VEGF minicircle DNA under control of the SV40 promoter (pMini-SV-VEGF) showed an increased amount of VEGF mRNA and up to 8 times more VEGF expression than a conventional plasmid (pSV-VEGF) in two different skeletal muscle cell lines (C2C12 and L8). Minicircle DNA with different promoters, including the SV40, CMV and chicken β-actin, was tested for VEGF expression in a C2C12 skeletal muscle cell line. The high VEGF expression generated by minicircle DNA stimulated efficient endothelial cell growth in vitro. Furthermore, minicircle DNA showed a higher VEGF expression compared to conventional plasmid in the tibialis anterior muscle of mice. These results suggest that minicircle DNA is an effective gene vector for angiogenic VEGF expression in skeletal muscle and may be useful for treating peripheral arterial disease.

10. Human EPO Gene Delivery

EPO plays a key regulatory role in the formation of new red blood cells (RBCs). EPO may also have a role as a therapeutic agent to counteract ischemic injury in neural, cardiac and endothelial cells [130]. Several reports have demonstrated the capacity of EPO to protect and revascularize the myocardium following ischemic injury [131-133]. One of the limitations preventing the therapeutic application of EPO is its short half-life. The development of a more sustained form of EPO with a longer half-life would remove a significant barrier to its development as a therapeutic agent. Recently, several researchers have focused on the development of an EPO plasmid DNA delivered using either viral or non-viral carriers to promote the prolonged and controlled release of EPO in vivo [134]. Despite widespread use of recombinant human EPO (rHuEPO), several clinical limitations remain, including frequent injections, limited routes of administration, high medical expenditures, development of autoimmune pure red cell aplasia, and impacts on hemoglobin variability [135, 136]. To overcome many of these clinical hurdles, gene therapy providing continuous release has been suggested as an attractive alternative to current intermittently administered erythropoiesis-stimulating agents (ESAs) [137]. The transfection assays of the pEPO polyplex have been carried out in a variety of cell types and the time-course release of EPO has been analyzed in vitro [138].

Beyond the conventional effect of secreted erythropoietin from the kidney in response to hypoxic stimuli, EPO was recently identified as a pleiotropic and organ-protective cytokine, mediating repair and regeneration via anti-apoptosis, anti-inflammation, anti-oxidation, pro-angiogenesis and reendothelialization, vascular-protectant, mobilization of endothelial progenitor cells, and recruitment of stem cells into the zone of damage [132]. Apart from traditional erythropoietic effects, the pleiotropic organ-protective effects of EPO make it a frontline cardioprotective candidate. Higher levels of endogenous EPO have been shown to have protective effects against I/R injury in acute MI in humans [139]. Along with numerous ex vivo and in vivo studies, some clinical studies with a single rHuEPO administration after the percutaneous coronary intervention showed favorable effects on infarct size, cardiac function, and patient prognosis [132, 140]. However, even though the in vitro and in vivo data supporting a rHuEPO cardioprotective approach are numerous, recent randomized clinical trials in acute MI patients have reported conflicting data [141]. To date, little is known about how polymer-mediated phEPO therapy distinctly alters cardiac remodeling in the rat MI model. The sustained release of intramyocardial phEPO gene therapy delivered by ABP polymer may restore heart function and limit pathological cardiac remodeling after MI [142]. Additionally, this finding assessed the effect of phEPO/ABP gene therapy on cardiac remodeling by evaluating the pro-fibrotic angiotensin II (Ang II) and TGF-β expression in rat hearts. Intramyocardial EPO gene therapy delivered by the bioreducible ABP polymer demonstrates potential cardioprotective effects on post-infarct cardiac remodeling in rats, compared with treatment of rHuEPO protein and naked phEPO plasmid-alone. The prominent effects of EPO/ABP gene therapy are accompanied by the preservation of cardiac geometry and function, reduction in the density of fibrotic tissue, protection against cardiomyocyte loss, decrease in apoptotic activity, stimulation of angiogenesis, inhibition of α-SMA+ myoFb differentiation, and suppression of the profibrotic Ang II and TGF-β expression across the LVfb and remote non-infarcted sites in rats after MI.

11. Conclusion

Inter-disciplinary researchers have advanced gene therapy due to its potential advantages over conventional treatment for myocardial ischemic diseases. Angiogenesis-based therapy using angiogenic genes have showed positive effects on the ischemic heart in preclinical and clinical studies. Anti apoptotic genes have also demonstrated the ability to prevent apoptosis-induced cell death by the inhibition of the apoptotic pathway in the ischemic heart. The intracellular delivery of anti-apoptotic genes needs improvement in order to increase the therapeutic efficacy and specificity to the targeted organ. Polymeric gene carries have been developed to substitute viral vectors for their avoidance of immunogenicity and oncogenecity. Although considerable improvements in gene transfer techniques using non-viral vectors have been witnessed during the last decade, the standard requirements for clinical use have not been met in terms of efficiency and specificity. When designing polymeric vectors, studies to overcome subcellular barriers including endosomal escape and nuclear translocation should be considered. Gene delivery is a multi-step process, which needs the appropriate carriers for each stage. Therefore, plasmid and multi functional polymeric vectors should be rationally designed. The plasmid design should include the introduction of tissue-specific promoters or regulatory promoters. As for the multifunctional polymeric vectors, they should be designed in conjunction with the vector development at the molecular level, taking into account the need to overcome a series of extra- and intra-cellular barriers.

Acknowledgments

This work was supported by the NIH grants HL06547

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Gheorghiade M, Bonow RO. Chronic heart failure in the United States: a manifestation of coronary artery disease. Circulation. 1998;97:282–289. doi: 10.1161/01.cir.97.3.282. [DOI] [PubMed] [Google Scholar]
  • 2.Wang XJ, Li QP. The roles of mesenchymal stem cells (MSCs) therapy in ischemic heart diseases. Biochemical and biophysical research communications. 2007;359:189–193. doi: 10.1016/j.bbrc.2007.05.112. [DOI] [PubMed] [Google Scholar]
  • 3.McGinn AN, Nam HY, Ou M, Hu N, Straub CM, Yockman JW, Bull DA, Kim SW. Bioreducible polymer-transfected skeletal myoblasts for VEGF delivery to acutely ischemic myocardium. Biomaterials. 2011;32:942–949. doi: 10.1016/j.biomaterials.2010.09.061. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Struthers AD. Pathophysiology of heart failure following myocardial infarction. Heart. 2005;91(Suppl 2):ii14–16. doi: 10.1136/hrt.2005.062034. discussion ii31, ii43-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Won YW, Kim JK, Cha MJ, Hwang KC, Choi D, Kim YH. Prolongation and enhancement of the anti-apoptotic effects of PTD-Hsp27 fusion proteins using an injectable thermo-reversible gel in a rat myocardial infarction model. Journal of controlled release : official journal of the Controlled Release Society. 2010;144:181–189. doi: 10.1016/j.jconrel.2010.02.014. [DOI] [PubMed] [Google Scholar]
  • 6.Ledley FD, McNamee LM, Uzdil V, Morgan IW. Why commercialization of gene therapy stalled; examining the life cycles of gene therapy technologies. Gene therapy. 2014;21:188–194. doi: 10.1038/gt.2013.72. [DOI] [PubMed] [Google Scholar]
  • 7.Whitehead KA, Langer R, Anderson DG. Knocking down barriers: advances in siRNA delivery. Nature reviews Drug discovery. 2009;8:129–138. doi: 10.1038/nrd2742. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Park TG, Jeong JH, Kim SW. Current status of polymeric gene delivery systems. Advanced drug delivery reviews. 2006;58:467–486. doi: 10.1016/j.addr.2006.03.007. [DOI] [PubMed] [Google Scholar]
  • 9.Kim SW. Biomaterials to gene delivery. Journal of controlled release : official journal of the Controlled Release Society. 2011;155:116–118. doi: 10.1016/j.jconrel.2011.03.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Kim WJ, Kim SW. Efficient siRNA delivery with non-viral polymeric vehicles. Pharmaceutical research. 2009;26:657–666. doi: 10.1007/s11095-008-9774-1. [DOI] [PubMed] [Google Scholar]
  • 11.Mack CA, Patel SR, Schwarz EA, Zanzonico P, Hahn RT, Ilercil A, Devereux RB, Goldsmith SJ, Christian TF, Sanborn TA, Kovesdi I, Hackett N, Isom OW, Crystal RG, Rosengart TK. Biologic bypass with the use of adenovirus-mediated gene transfer of the complementary deoxyribonucleic acid for vascular endothelial growth factor 121 improves myocardial perfusion and function in the ischemic porcine heart. The Journal of thoracic and cardiovascular surgery. 1998;115:168–176. doi: 10.1016/s0022-5223(98)70455-6. discussion 176-167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Giordano FJ, Ping P, McKirnan MD, Nozaki S, DeMaria AN, Dillmann WH, Mathieu-Costello O, Hammond HK. Intracoronary gene transfer of fibroblast growth factor-5 increases blood flow and contractile function in an ischemic region of the heart. Nature medicine. 1996;2:534–539. doi: 10.1038/nm0596-534. [DOI] [PubMed] [Google Scholar]
  • 13.Losordo DW, Vale PR, Hendel RC, Milliken CE, Fortuin FD, Cummings N, Schatz RA, Asahara T, Isner JM, Kuntz RE. Phase 1/2 placebo-controlled, double-blind, dose-escalating trial of myocardial vascular endothelial growth factor 2 gene transfer by catheter delivery in patients with chronic myocardial ischemia. Circulation. 2002;105:2012–2018. doi: 10.1161/01.cir.0000015982.70785.b7. [DOI] [PubMed] [Google Scholar]
  • 14.Laham RJ, Sellke FW, Edelman ER, Pearlman JD, Ware JA, Brown DL, Gold JP, Simons M. Local perivascular delivery of basic fibroblast growth factor in patients undergoing coronary bypass surgery: results of a phase I randomized, double-blind, placebo-controlled trial. Circulation. 1999;100:1865–1871. doi: 10.1161/01.cir.100.18.1865. [DOI] [PubMed] [Google Scholar]
  • 15.Akhtar S, Hughes MD, Khan A, Bibby M, Hussain M, Nawaz Q, Double J, Sayyed P. The delivery of antisense therapeutics. Advanced drug delivery reviews. 2000;44:3–21. doi: 10.1016/s0169-409x(00)00080-6. [DOI] [PubMed] [Google Scholar]
  • 16.Gilmore IR, Fox SP, Hollins AJ, Sohail M, Akhtar S. The design and exogenous delivery of siRNA for post-transcriptional gene silencing. Journal of drug targeting. 2004;12:315–340. doi: 10.1080/10611860400006257. [DOI] [PubMed] [Google Scholar]
  • 17.Hughes MD, Hussain M, Nawaz Q, Sayyed P, Akhtar S. The cellular delivery of antisense oligonucleotides and ribozymes. Drug discovery today. 2001;6:303–315. doi: 10.1016/s1359-6446(00)00326-3. [DOI] [PubMed] [Google Scholar]
  • 18.Ahn CH, Chae SY, Bae YH, Kim SW. Biodegradable poly(ethylenimine) for plasmid DNA delivery. Journal of controlled release : official journal of the Controlled Release Society. 2002;80:273–282. doi: 10.1016/s0168-3659(01)00547-8. [DOI] [PubMed] [Google Scholar]
  • 19.Kim TI, Rothmund T, Kissel T, Kim SW. Bioreducible polymers with cell penetrating and endosome buffering functionality for gene delivery systems. Journal of controlled release : official journal of the Controlled Release Society. 2011;152:110–119. doi: 10.1016/j.jconrel.2011.02.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Ou M, Xu R, Kim SH, Bull DA, Kim SW. A family of bioreducible poly(disulfide amine)s for gene delivery. Biomaterials. 2009;30:5804–5814. doi: 10.1016/j.biomaterials.2009.06.050. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Won YW, Lim KS, Kim YH. Intracellular organelle-targeted non-viral gene delivery systems. Journal of controlled release : official journal of the Controlled Release Society. 2011;152:99–109. doi: 10.1016/j.jconrel.2011.01.013. [DOI] [PubMed] [Google Scholar]
  • 22.Kim JS, Maruyama A, Akaike T, Kim SW. Terplex DNA delivery system as a gene carrier. Pharmaceutical research. 1998;15:116–121. doi: 10.1023/a:1011917224044. [DOI] [PubMed] [Google Scholar]
  • 23.Kim JS, Kim BI, Maruyama A, Akaike T, Kim SW. A new non-viral DNA delivery vector: the terplex system. Journal of controlled release : official journal of the Controlled Release Society. 1998;53:175–182. doi: 10.1016/s0168-3659(97)00251-4. [DOI] [PubMed] [Google Scholar]
  • 24.Yu L, Nielsen M, Han SO, Wan Kim S. TerplexDNA gene carrier system targeting artery wall cells. Journal of controlled release : official journal of the Controlled Release Society. 2001;72:179–189. doi: 10.1016/s0168-3659(01)00274-7. [DOI] [PubMed] [Google Scholar]
  • 25.Sawamura T, Kume N, Aoyama T, Moriwaki H, Hoshikawa H, Aiba Y, Tanaka T, Miwa S, Katsura Y, Kita T, Masaki T. An endothelial receptor for oxidized low-density lipoprotein. Nature. 1997;386:73–77. doi: 10.1038/386073a0. [DOI] [PubMed] [Google Scholar]
  • 26.Shih IL, Lees RS, Chang MY, Lees AM. Focal accumulation of an apolipoprotein B-based synthetic oligopeptide in the healing rabbit arterial wall. Proceedings of the National Academy of Sciences of the United States of America. 1990;87:1436–1440. doi: 10.1073/pnas.87.4.1436. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Lestavel-Delattre S, Martin-Nizard F, Clavey V, Testard P, Favre G, Doualin G, Houssaini HS, Bard JM, Duriez P, Delbart C, et al. Low-density lipoprotein for delivery of an acrylophenone antineoplastic molecule into malignant cells. Cancer research. 1992;52:3629–3635. [PubMed] [Google Scholar]
  • 28.Affleck DG, Yu L, Bull DA, Bailey SH, Kim SW. Augmentation of myocardial transfection using TerplexDNA: a novel gene delivery system. Gene therapy. 2001;8:349–353. doi: 10.1038/sj.gt.3301395. [DOI] [PubMed] [Google Scholar]
  • 29.Sutton MG, Sharpe N. Left ventricular remodeling after myocardial infarction: pathophysiology and therapy. Circulation. 2000;101:2981–2988. doi: 10.1161/01.cir.101.25.2981. [DOI] [PubMed] [Google Scholar]
  • 30.Giordano FJ, Gerber HP, Williams SP, VanBruggen N, Bunting S, Ruiz-Lozano P, Gu Y, Nath AK, Huang Y, Hickey R, Dalton N, Peterson KL, Ross J, Jr, Chien KR, Ferrara N. A cardiac myocyte vascular endothelial growth factor paracrine pathway is required to maintain cardiac function. Proceedings of the National Academy of Sciences of the United States of America. 2001;98:5780–5785. doi: 10.1073/pnas.091415198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Luo Z, Diaco M, Murohara T, Ferrara N, Isner JM, Symes JF. Vascular endothelial growth factor attenuates myocardial ischemia-reperfusion injury. The Annals of thoracic surgery. 1997;64:993–998. doi: 10.1016/s0003-4975(97)00715-7. [DOI] [PubMed] [Google Scholar]
  • 32.Bull DA, Bailey SH, Rentz JJ, Zebrack JS, Lee M, Litwin SE, Kim SW. Effect of Terplex/VEGF-165 gene therapy on left ventricular function and structure following myocardial infarction. VEGF gene therapy for myocardial infarction. Journal of controlled release : official journal of the Controlled Release Society. 2003;93:175–181. doi: 10.1016/j.jconrel.2003.06.002. [DOI] [PubMed] [Google Scholar]
  • 33.Han S, Mahato RI, Kim SW. Water-soluble lipopolymer for gene delivery. Bioconjugate chemistry. 2001;12:337–345. doi: 10.1021/bc000120w. [DOI] [PubMed] [Google Scholar]
  • 34.Lee M, Rentz J, Han SO, Bull DA, Kim SW. Water-soluble lipopolymer as an efficient carrier for gene delivery to myocardium. Gene therapy. 2003;10:585–593. doi: 10.1038/sj.gt.3301938. [DOI] [PubMed] [Google Scholar]
  • 35.Lee M, Rentz J, Bikram M, Han S, Bull DA, Kim SW. Hypoxia-inducible VEGF gene delivery to ischemic myocardium using water-soluble lipopolymer. Gene therapy. 2003;10:1535–1542. doi: 10.1038/sj.gt.3302034. [DOI] [PubMed] [Google Scholar]
  • 36.Benns JM, Maheshwari A, Furgeson DY, Mahato RI, Kim SW. Folate-PEG-folate-graft-polyethylenimine-based gene delivery. Journal of drug targeting. 2001;9:123–139. doi: 10.3109/10611860108997923. [DOI] [PubMed] [Google Scholar]
  • 37.Simons K, Ikonen E. How cells handle cholesterol. Science. 2000;290:1721–1726. doi: 10.1126/science.290.5497.1721. [DOI] [PubMed] [Google Scholar]
  • 38.Lee M, Han S, Ko KS, Kim SW. Cell type specific and glucose responsive expression of interleukin-4 by using insulin promoter and water soluble lipopolymer. Journal of controlled release : official journal of the Controlled Release Society. 2001;75:421–429. doi: 10.1016/s0168-3659(01)00416-3. [DOI] [PubMed] [Google Scholar]
  • 39.Isner JM. Myocardial gene therapy. Nature. 2002;415:234–239. doi: 10.1038/415234a. [DOI] [PubMed] [Google Scholar]
  • 40.Springer ML, Chen AS, Kraft PE, Bednarski M, Blau HM. VEGF gene delivery to muscle: potential role for vasculogenesis in adults. Molecular cell. 1998;2:549–558. doi: 10.1016/s1097-2765(00)80154-9. [DOI] [PubMed] [Google Scholar]
  • 41.Lee RJ, Springer ML, Blanco-Bose WE, Shaw R, Ursell PC, Blau HM. VEGF gene delivery to myocardium: deleterious effects of unregulated expression. Circulation. 2000;102:898–901. doi: 10.1161/01.cir.102.8.898. [DOI] [PubMed] [Google Scholar]
  • 42.Maxwell PH, Pugh CW, Ratcliffe PJ. Inducible operation of the erythropoietin 3′ enhancer in multiple cell lines: evidence for a widespread oxygen-sensing mechanism. Proceedings of the National Academy of Sciences of the United States of America. 1993;90:2423–2427. doi: 10.1073/pnas.90.6.2423. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Jiang BH, Rue E, Wang GL, Roe R, Semenza GL. Dimerization, DNA binding, and transactivation properties of hypoxia-inducible factor 1. The Journal of biological chemistry. 1996;271:17771–17778. doi: 10.1074/jbc.271.30.17771. [DOI] [PubMed] [Google Scholar]
  • 44.Wang GL, Semenza GL. General involvement of hypoxia-inducible factor 1 in transcriptional response to hypoxia. Proceedings of the National Academy of Sciences of the United States of America. 1993;90:4304–4308. doi: 10.1073/pnas.90.9.4304. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Levy AP, Levy NS, Wegner S, Goldberg MA. Transcriptional regulation of the rat vascular endothelial growth factor gene by hypoxia. The Journal of biological chemistry. 1995;270:13333–13340. doi: 10.1074/jbc.270.22.13333. [DOI] [PubMed] [Google Scholar]
  • 46.Liu Y, Cox SR, Morita T, Kourembanas S. Hypoxia regulates vascular endothelial growth factor gene expression in endothelial cells. Identification of a 5′ enhancer. Circulation research. 1995;77:638–643. doi: 10.1161/01.res.77.3.638. [DOI] [PubMed] [Google Scholar]
  • 47.Forsythe JA, Jiang BH, Iyer NV, Agani F, Leung SW, Koos RD, Semenza GL. Activation of vascular endothelial growth factor gene transcription by hypoxia-inducible factor 1. Molecular and cellular biology. 1996;16:4604–4613. doi: 10.1128/mcb.16.9.4604. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Stein I, Neeman M, Shweiki D, Itin A, Keshet E. Stabilization of vascular endothelial growth factor mRNA by hypoxia and hypoglycemia and coregulation with other ischemia-induced genes. Molecular and cellular biology. 1995;15:5363–5368. doi: 10.1128/mcb.15.10.5363. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Su H, Arakawa-Hoyt J, Kan YW. Adeno-associated viral vector-mediated hypoxia response element-regulated gene expression in mouse ischemic heart model. Proceedings of the National Academy of Sciences of the United States of America. 2002;99:9480–9485. doi: 10.1073/pnas.132275299. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Shoshani T, Faerman A, Mett I, Zelin E, Tenne T, Gorodin S, Moshel Y, Elbaz S, Budanov A, Chajut A, Kalinski H, Kamer I, Rozen A, Mor O, Keshet E, Leshkowitz D, Einat P, Skaliter R, Feinstein E. Identification of a novel hypoxia-inducible factor 1-responsive gene, RTP801, involved in apoptosis. Molecular and cellular biology. 2002;22:2283–2293. doi: 10.1128/MCB.22.7.2283-2293.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Wang GL, Jiang BH, Rue EA, Semenza GL. Hypoxia-inducible factor 1 is a basic-helix-loop-helix-PAS heterodimer regulated by cellular O2 tension. Proceedings of the National Academy of Sciences of the United States of America. 1995;92:5510–5514. doi: 10.1073/pnas.92.12.5510. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Wenger RH, Gassmann M. Oxygen(es) and the hypoxia-inducible factor-1. Biological chemistry. 1997;378:609–616. [PubMed] [Google Scholar]
  • 53.Semenza GL, Jiang BH, Leung SW, Passantino R, Concordet JP, Maire P, Giallongo A. Hypoxia response elements in the aldolase A, enolase 1, and lactate dehydrogenase A gene promoters contain essential binding sites for hypoxia-inducible factor 1. The Journal of biological chemistry. 1996;271:32529–32537. doi: 10.1074/jbc.271.51.32529. [DOI] [PubMed] [Google Scholar]
  • 54.Lee M, Bikram M, Oh S, Bull DA, Kim SW. Sp1-dependent regulation of the RTP801 promoter and its application to hypoxia-inducible VEGF plasmid for ischemic disease. Pharmaceutical research. 2004;21:736–741. doi: 10.1023/b:pham.0000026421.09367.b3. [DOI] [PubMed] [Google Scholar]
  • 55.Christensen LV, Chang CW, Yockman JW, Conners R, Jackson H, Zhong Z, Feijen J, Bull DA, Kim SW. Reducible poly(amido ethylenediamine) for hypoxia-inducible VEGF delivery. Journal of controlled release : official journal of the Controlled Release Society. 2007;118:254–261. doi: 10.1016/j.jconrel.2006.12.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Crottogini A, Meckert PC, Vera Janavel G, Lascano E, Negroni J, Del Valle H, Dulbecco E, Werba P, Cuniberti L, Martinez V, De Lorenzi A, Telayna J, Mele A, Fernandez JL, Marangunich L, Criscuolo M, Capogrossi MC, Laguens R. Arteriogenesis induced by intramyocardial vascular endothelial growth factor 165 gene transfer in chronically ischemic pigs. Human gene therapy. 2003;14:1307–1318. doi: 10.1089/104303403322319390. [DOI] [PubMed] [Google Scholar]
  • 57.Laguens R, Cabeza Meckert P, Vera Janavel G, De Lorenzi A, Lascano E, Negroni J, Del Valle H, Cuniberti L, Martinez V, Dulbecco E, Melo C, Fernandez N, Criscuolo M, Crottogini A. Cardiomyocyte hyperplasia after plasmid-mediated vascular endothelial growth factor gene transfer in pigs with chronic myocardial ischemia. The journal of gene medicine. 2004;6:222–227. doi: 10.1002/jgm.478. [DOI] [PubMed] [Google Scholar]
  • 58.Vera Janavel G, Crottogini A, Cabeza Meckert P, Cuniberti L, Mele A, Papouchado M, Fernandez N, Bercovich A, Criscuolo M, Melo C, Laguens R. Plasmid-mediated VEGF gene transfer induces cardiomyogenesis and reduces myocardial infarct size in sheep. Gene therapy. 2006;13:1133–1142. doi: 10.1038/sj.gt.3302708. [DOI] [PubMed] [Google Scholar]
  • 59.Lee S, Kim K, Kim HA, Kim SW, Lee M. Augmentation of erythropoietin enhancer-mediated hypoxia-inducible gene expression by co-transfection of a plasmid encoding hypoxia-inducible factor 1alpha for ischemic tissue targeting gene therapy. Journal of drug targeting. 2008;16:43–50. doi: 10.1080/10611860701699693. [DOI] [PubMed] [Google Scholar]
  • 60.Kim HA, Mahato RI, Lee M. Hypoxia-specific gene expression for ischemic disease gene therapy. Advanced drug delivery reviews. 2009;61:614–622. doi: 10.1016/j.addr.2009.04.009. [DOI] [PubMed] [Google Scholar]
  • 61.Kim HA, Lim S, Moon HH, Kim SW, Hwang KC, Lee M, Kim SH, Choi D. Hypoxia-inducible vascular endothelial growth factor gene therapy using the oxygen-dependent degradation domain in myocardial ischemia. Pharmaceutical research. 2010;27:2075–2084. doi: 10.1007/s11095-010-0206-7. [DOI] [PubMed] [Google Scholar]
  • 62.Jin H, Liu ML, Kim HA, Lee M, An S, Oh J, Cho J, Yi S, Kim K, Yoon D, Ha Y. Role of the oxygen-dependent degradation domain in a hypoxia-inducible gene expression system in vascular endothelial growth factor gene therapy. Spine. 2009;34:E952–958. doi: 10.1097/BRS.0b013e3181c4af80. [DOI] [PubMed] [Google Scholar]
  • 63.Stefanini MO, Wu FT, Mac Gabhann F, Popel AS. A compartment model of VEGF distribution in blood, healthy and diseased tissues. BMC systems biology. 2008;2:77. doi: 10.1186/1752-0509-2-77. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.George ML, Eccles SA, Tutton MG, Abulafi AM, Swift RI. Correlation of plasma and serum vascular endothelial growth factor levels with platelet count in colorectal cancer: clinical evidence of platelet scavenging? Clinical cancer research : an official journal of the American Association for Cancer Research. 2000;6:3147–3152. [PubMed] [Google Scholar]
  • 65.Rudge JS, Holash J, Hylton D, Russell M, Jiang S, Leidich R, Papadopoulos N, Pyles EA, Torri A, Wiegand SJ, Thurston G, Stahl N, Yancopoulos GD. VEGF Trap complex formation measures production rates of VEGF, providing a biomarker for predicting efficacious angiogenic blockade. Proceedings of the National Academy of Sciences of the United States of America. 2007;104:18363–18370. doi: 10.1073/pnas.0708865104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Won YW, Lee M, Kim HA, Bull DA, Kim SW. Post-translational regulated and hypoxia-responsible VEGF plasmid for efficient secretion. Journal of controlled release : official journal of the Controlled Release Society. 2012;160:525–531. doi: 10.1016/j.jconrel.2012.03.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Koditz J, Nesper J, Wottawa M, Stiehl DP, Camenisch G, Franke C, Myllyharju J, Wenger RH, Katschinski DM. Oxygen-dependent ATF-4 stability is mediated by the PHD3 oxygen sensor. Blood. 2007;110:3610–3617. doi: 10.1182/blood-2007-06-094441. [DOI] [PubMed] [Google Scholar]
  • 68.Kim HA, Kim K, Kim SW, Lee M. Transcriptional and post-translational regulatory system for hypoxia specific gene expression using the erythropoietin enhancer and the oxygen-dependent degradation domain. Journal of controlled release : official journal of the Controlled Release Society. 2007;121:218–224. doi: 10.1016/j.jconrel.2007.05.036. [DOI] [PubMed] [Google Scholar]
  • 69.Harada H, Hiraoka M, Kizaka-Kondoh S. Antitumor effect of TAT-oxygen-dependent degradationcaspase-3 fusion protein specifically stabilized and activated in hypoxic tumor cells. Cancer research. 2002;62:2013–2018. [PubMed] [Google Scholar]
  • 70.Kim TI, Lee M, Kim SW. Efficient GLP-1 gene delivery using two-step transcription amplification plasmid system with a secretion signal peptide and arginine-grafted bioreducible polymer. Journal of controlled release : official journal of the Controlled Release Society. 2012;157:243–248. doi: 10.1016/j.jconrel.2011.09.072. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Thomas G. Furin at the cutting edge: from protein traffic to embryogenesis and disease, Nature reviews. Molecular cell biology. 2002;3:753–766. doi: 10.1038/nrm934. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Molloy SS, Bresnahan PA, Leppla SH, Klimpel KR, Thomas G. Human furin is a calcium-dependent serine endoprotease that recognizes the sequence Arg-X-X-Arg and efficiently cleaves anthrax toxin protective antigen. The Journal of biological chemistry. 1992;267:16396–16402. [PubMed] [Google Scholar]
  • 73.Walker JA, Molloy SS, Thomas G, Sakaguchi T, Yoshida T, Chambers TM, Kawaoka Y. Sequence specificity of furin, a proprotein-processing endoprotease, for the hemagglutinin of a virulent avian influenza virus. Journal of virology. 1994;68:1213–1218. doi: 10.1128/jvi.68.2.1213-1218.1994. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Oh S, Lee M, Ko KS, Choi S, Kim SW. GLP-1 gene delivery for the treatment of type 2 diabetes. Molecular therapy : the journal of the American Society of Gene Therapy. 2003;7:478–483. doi: 10.1016/s1525-0016(03)00036-4. [DOI] [PubMed] [Google Scholar]
  • 75.Dai Y, Xu M, Wang Y, Pasha Z, Li T, Ashraf M. HIF-1alpha induced-VEGF overexpression in bone marrow stem cells protects cardiomyocytes against ischemia. Journal of molecular and cellular cardiology. 2007;42:1036–1044. doi: 10.1016/j.yjmcc.2007.04.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Gupta K, Kshirsagar S, Li W, Gui L, Ramakrishnan S, Gupta P, Law PY, Hebbel RP. VEGF prevents apoptosis of human microvascular endothelial cells via opposing effects on MAPK/ERK and SAPK/JNK signaling. Experimental cell research. 1999;247:495–504. doi: 10.1006/excr.1998.4359. [DOI] [PubMed] [Google Scholar]
  • 77.Xu Q, Ji YS, Schmedtje JF., Jr Sp1 increases expression of cyclooxygenase-2 in hypoxic vascular endothelium. Implications for the mechanisms of aortic aneurysm and heart failure. The Journal of biological chemistry. 2000;275:24583–24589. doi: 10.1074/jbc.M003894200. [DOI] [PubMed] [Google Scholar]
  • 78.Won YW, McGinn AN, Lee M, Nam K, Bull DA, Kim SW. Post-translational regulation of a hypoxia-responsive VEGF plasmid for the treatment of myocardial ischemia. Biomaterials. 2013;34:6229–6238. doi: 10.1016/j.biomaterials.2013.04.061. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Vinge LE, Raake PW, Koch WJ. Gene therapy in heart failure. Circulation research. 2008;102:1458–1470. doi: 10.1161/CIRCRESAHA.108.173195. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Lin HH, Chen YH, Chang PF, Lee YT, Yet SF, Chau LY. Heme oxygenase-1 promotes neovascularization in ischemic heart by coinduction of VEGF and SDF-1. Journal of molecular and cellular cardiology. 2008;45:44–55. doi: 10.1016/j.yjmcc.2008.04.011. [DOI] [PubMed] [Google Scholar]
  • 81.Suzuki M, Iso-o N, Takeshita S, Tsukamoto K, Mori I, Sato T, Ohno M, Nagai R, Ishizaka N. Facilitated angiogenesis induced by heme oxygenase-1 gene transfer in a rat model of hindlimb ischemia. Biochemical and biophysical research communications. 2003;302:138–143. doi: 10.1016/s0006-291x(03)00114-1. [DOI] [PubMed] [Google Scholar]
  • 82.Sugano M, Tsuchida K, Maeda T, Makino N. SiRNA targeting SHP-1 accelerates angiogenesis in a rat model of hindlimb ischemia. Atherosclerosis. 2007;191:33–39. doi: 10.1016/j.atherosclerosis.2006.04.021. [DOI] [PubMed] [Google Scholar]
  • 83.Stocker R, Perrella MA. Heme oxygenase-1: a novel drug target for atherosclerotic diseases? Circulation. 2006;114:2178–2189. doi: 10.1161/CIRCULATIONAHA.105.598698. [DOI] [PubMed] [Google Scholar]
  • 84.Tang YL, Tang Y, Zhang YC, Qian K, Shen L, Phillips MI. Protection from ischemic heart injury by a vigilant heme oxygenase-1 plasmid system. Hypertension. 2004;43:746–751. doi: 10.1161/01.HYP.0000120152.27263.87. [DOI] [PubMed] [Google Scholar]
  • 85.Chong ZZ, Maiese K. The Src homology 2 domain tyrosine phosphatases SHP-1 and SHP-2: diversified control of cell growth, inflammation, and injury. Histology and histopathology. 2007;22:1251–1267. doi: 10.14670/hh-22.1251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Forget G, Gregory DJ, Whitcombe LA, Olivier M. Role of host protein tyrosine phosphatase SHP-1 in Leishmania donovani-induced inhibition of nitric oxide production. Infection and immunity. 2006;74:6272–6279. doi: 10.1128/IAI.00853-05. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Matsui T, Tao J, del Monte F, Lee KH, Li L, Picard M, Force TL, Franke TF, Hajjar RJ, Rosenzweig A. Akt activation preserves cardiac function and prevents injury after transient cardiac ischemia in vivo. Circulation. 2001;104:330–335. doi: 10.1161/01.cir.104.3.330. [DOI] [PubMed] [Google Scholar]
  • 88.Nam HY, Kim J, Kim S, Yockman JW, Kim SW, Bull DA. Cell penetrating peptide conjugated bioreducible polymer for siRNA delivery. Biomaterials. 2011;32:5213–5222. doi: 10.1016/j.biomaterials.2011.03.058. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Lee Y, Jeon K, Lee JT, Kim S, Kim VN. MicroRNA maturation: stepwise processing and subcellular localization. The EMBO journal. 2002;21:4663–4670. doi: 10.1093/emboj/cdf476. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Won YW, Lee M, Kim HA, Bull DA, Kim SW. Hypoxia-inducible plasmid expressing both miSHP-1 and HO-1 for the treatment of ischemic disease. Journal of controlled release : official journal of the Controlled Release Society. 2013;165:22–28. doi: 10.1016/j.jconrel.2012.10.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Won YW, Lee M, Kim HA, Nam K, Bull DA, Kim SW. Synergistically combined gene delivery for enhanced VEGF secretion and antiapoptosis. Molecular pharmaceutics. 2013;10:3676–3683. doi: 10.1021/mp400178m. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.May D, Gilon D, Djonov V, Itin A, Lazarus A, Gordon O, Rosenberger C, Keshet E. Transgenic system for conditional induction and rescue of chronic myocardial hibernation provides insights into genomic programs of hibernation. Proceedings of the National Academy of Sciences of the United States of America. 2008;105:282–287. doi: 10.1073/pnas.0707778105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Nah JW, Yu L, Han SO, Ahn CH, Kim SW. Artery wall binding peptide-poly(ethylene glycol)-grafted-poly(L-lysine)-based gene delivery to artery wall cells. Journal of controlled release : official journal of the Controlled Release Society. 2002;78:273–284. doi: 10.1016/s0168-3659(01)00499-0. [DOI] [PubMed] [Google Scholar]
  • 94.Kawakami S, Sato A, Nishikawa M, Yamashita F, Hashida M. Mannose receptor-mediated gene transfer into macrophages using novel mannosylated cationic liposomes. Gene therapy. 2000;7:292–299. doi: 10.1038/sj.gt.3301089. [DOI] [PubMed] [Google Scholar]
  • 95.Hwa Kim S, Hoon Jeong J, Chul Cho K, Wan Kim S, Gwan Park T. Target-specific gene silencing by siRNA plasmid DNA complexed with folate-modified poly(ethylenimine) Journal of controlled release : official journal of the Controlled Release Society. 2005;104:223–232. doi: 10.1016/j.jconrel.2005.02.006. [DOI] [PubMed] [Google Scholar]
  • 96.Sato A, Takagi M, Shimamoto A, Kawakami S, Hashida M. Small interfering RNA delivery to the liver by intravenous administration of galactosylated cationic liposomes in mice. Biomaterials. 2007;28:1434–1442. doi: 10.1016/j.biomaterials.2006.11.010. [DOI] [PubMed] [Google Scholar]
  • 97.Bergstrom S, Carlson LA, Weeks JR. The prostaglandins: a family of biologically active lipids. Pharmacological reviews. 1968;20:1–48. [PubMed] [Google Scholar]
  • 98.Coleman RA, Smith WL, Narumiya S. International Union of Pharmacology classification of prostanoid receptors: properties, distribution, and structure of the receptors and their subtypes. Pharmacological reviews. 1994;46:205–229. [PubMed] [Google Scholar]
  • 99.Desai S, April H, Nwaneshiudu C, Ashby B. Comparison of agonist-induced internalization of the human EP2 and EP4 prostaglandin receptors: role of the carboxyl terminus in EP4 receptor sequestration. Molecular pharmacology. 2000;58:1279–1286. doi: 10.1124/mol.58.6.1279. [DOI] [PubMed] [Google Scholar]
  • 100.Xiao CY, Yuhki K, Hara A, Fujino T, Kuriyama S, Yamada T, Takayama K, Takahata O, Karibe H, Taniguchi T, Narumiya S, Ushikubi F. Prostaglandin E2 protects the heart from ischemia-reperfusion injury via its receptor subtype EP4. Circulation. 2004;109:2462–2468. doi: 10.1161/01.CIR.0000128046.54681.97. [DOI] [PubMed] [Google Scholar]
  • 101.Christensen LV, Chang CW, Kim WJ, Kim SW, Zhong Z, Lin C, Engbersen JF, Feijen J. Reducible poly(amido ethylenimine)s designed for triggered intracellular gene delivery. Bioconjugate chemistry. 2006;17:1233–1240. doi: 10.1021/bc0602026. [DOI] [PubMed] [Google Scholar]
  • 102.Hoon Jeong J, Christensen LV, Yockman JW, Zhong Z, Engbersen JF, Jong Kim W, Feijen J, Wan Kim S. Reducible poly(amido ethylenimine) directed to enhance RNA interference. Biomaterials. 2007;28:1912–1917. doi: 10.1016/j.biomaterials.2006.12.019. [DOI] [PubMed] [Google Scholar]
  • 103.Ou M, Wang XL, Xu R, Chang CW, Bull DA, Kim SW. Novel biodegradable poly(disulfide amine)s for gene delivery with high efficiency and low cytotoxicity. Bioconjugate chemistry. 2008;19:626–633. doi: 10.1021/bc700397x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Kim SH, Jeong JH, Ou M, Yockman JW, Kim SW, Bull DA. Cardiomyocyte-targeted siRNA delivery by prostaglandin E(2)-Fas siRNA polyplexes formulated with reducible poly(amido amine) for preventing cardiomyocyte apoptosis. Biomaterials. 2008;29:4439–4446. doi: 10.1016/j.biomaterials.2008.07.047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.McGuire MJ, Samli KN, Johnston SA, Brown KC. In vitro selection of a peptide with high selectivity for cardiomyocytes in vivo. Journal of molecular biology. 2004;342:171–182. doi: 10.1016/j.jmb.2004.06.029. [DOI] [PubMed] [Google Scholar]
  • 106.Barry MA, Dower WJ, Johnston SA. Toward cell-targeting gene therapy vectors: selection of cell-binding peptides from random peptide-presenting phage libraries. Nature medicine. 1996;2:299–305. doi: 10.1038/nm0396-299. [DOI] [PubMed] [Google Scholar]
  • 107.Hamar P, Song E, Kokeny G, Chen A, Ouyang N, Lieberman J. Small interfering RNA targeting Fas protects mice against renal ischemia-reperfusion injury. Proceedings of the National Academy of Sciences of the United States of America. 2004;101:14883–14888. doi: 10.1073/pnas.0406421101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Stephanou A, Scarabelli TM, Brar BK, Nakanishi Y, Matsumura M, Knight RA, Latchman DS. Induction of apoptosis and Fas receptor/Fas ligand expression by ischemia/reperfusion in cardiac myocytes requires serine 727 of the STAT-1 transcription factor but not tyrosine 701. The Journal of biological chemistry. 2001;276:28340–28347. doi: 10.1074/jbc.M101177200. [DOI] [PubMed] [Google Scholar]
  • 109.Wang J, Li W, Min J, Ou Q, Chen J. Fas siRNA reduces apoptotic cell death of allogeneictransplanted hepatocytes in mouse spleen. Transplantation proceedings. 2003;35:1594–1595. doi: 10.1016/s0041-1345(03)00438-x. [DOI] [PubMed] [Google Scholar]
  • 110.Nam HY, McGinn A, Kim PH, Kim SW, Bull DA. Primary cardiomyocyte-targeted bioreducible polymer for efficient gene delivery to the myocardium. Biomaterials. 2010;31:8081–8087. doi: 10.1016/j.biomaterials.2010.07.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Nam HY, Kim J, Kim SW, Bull DA. Cell targeting peptide conjugation to siRNA polyplexes for effective gene silencing in cardiomyocytes. Molecular pharmaceutics. 2012;9:1302–1309. doi: 10.1021/mp200589z. [DOI] [PubMed] [Google Scholar]
  • 112.Kanki S, Jaalouk DE, Lee S, Yu AY, Gannon J, Lee RT. Identification of targeting peptides for ischemic myocardium by in vivo phage display. Journal of molecular and cellular cardiology. 2011;50:841–848. doi: 10.1016/j.yjmcc.2011.02.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Pasqualini R, Ruoslahti E. Organ targeting in vivo using phage display peptide libraries. Nature. 1996;380:364–366. doi: 10.1038/380364a0. [DOI] [PubMed] [Google Scholar]
  • 114.Hajitou A, Pasqualini R, Arap W. Vascular targeting: recent advances and therapeutic perspectives. Trends in cardiovascular medicine. 2006;16:80–88. doi: 10.1016/j.tcm.2006.01.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Won YW, Kim HA, Lee M, Kim YH. Reducible poly(oligo-D-arginine) for enhanced gene expression in mouse lung by intratracheal injection. Molecular therapy : the journal of the American Society of Gene Therapy. 2010;18:734–742. doi: 10.1038/mt.2009.297. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Kim WJ, Christensen LV, Jo S, Yockman JW, Jeong JH, Kim YH, Kim SW. Cholesteryl oligoarginine delivering vascular endothelial growth factor siRNA effectively inhibits tumor growth in colon adenocarcinoma. Molecular therapy : the journal of the American Society of Gene Therapy. 2006;14:343–350. doi: 10.1016/j.ymthe.2006.03.022. [DOI] [PubMed] [Google Scholar]
  • 117.Won YW, Yoon SM, Lee KM, Kim YH. Poly(oligo-D-arginine) with internal disulfide linkages as a cytoplasm-sensitive carrier for siRNA delivery. Molecular therapy : the journal of the American Society of Gene Therapy. 2011;19:372–380. doi: 10.1038/mt.2010.242. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Muller OJ, Katus HA, Bekeredjian R. Targeting the heart with gene therapy-optimized gene delivery methods. Cardiovascular research. 2007;73:453–462. doi: 10.1016/j.cardiores.2006.09.021. [DOI] [PubMed] [Google Scholar]
  • 119.Won YW, McGinn AN, Lee M, Bull DA, Kim SW. Targeted gene delivery to ischemic myocardium by homing peptide-guided polymeric carrier. Molecular pharmaceutics. 2013;10:378–385. doi: 10.1021/mp300500y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Haider HK, Elmadbouh I, Jean-Baptiste M, Ashraf M. Nonviral vector gene modification of stem cells for myocardial repair. Molecular medicine. 2008;14:79–86. doi: 10.2119/2007-00092.Haider. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Marshall E. Gene therapy death prompts review of adenovirus vector. Science. 1999;286:2244–2245. doi: 10.1126/science.286.5448.2244. [DOI] [PubMed] [Google Scholar]
  • 122.Lehrman S. Virus treatment questioned after gene therapy death. Nature. 1999;401:517–518. doi: 10.1038/43977. [DOI] [PubMed] [Google Scholar]
  • 123.von Degenfeld G, Banfi A, Springer ML, Wagner RA, Jacobi J, Ozawa CR, Merchant MJ, Cooke JP, Blau HM. Microenvironmental VEGF distribution is critical for stable and functional vessel growth in ischemia. FASEB journal : official publication of the Federation of American Societies for Experimental Biology. 2006;20:2657–2659. doi: 10.1096/fj.06-6568fje. [DOI] [PubMed] [Google Scholar]
  • 124.Ou M, Kim TI, Yockman JW, Borden BA, Bull DA, Kim SW. Polymer transfected primary myoblasts mediated efficient gene expression and angiogenic proliferation. Journal of controlled release : official journal of the Controlled Release Society. 2010;142:61–69. doi: 10.1016/j.jconrel.2009.09.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.von Degenfeld G, Banfi A, Springer ML, Blau HM. Myoblast-mediated gene transfer for therapeutic angiogenesis and arteriogenesis. British journal of pharmacology. 2003;140:620–626. doi: 10.1038/sj.bjp.0705492. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Menasche P. Skeletal muscle satellite cell transplantation. Cardiovascular research. 2003;58:351–357. doi: 10.1016/s0008-6363(02)00769-1. [DOI] [PubMed] [Google Scholar]
  • 127.Yockman JW, Choi D, Whitten MG, Chang CW, Kastenmeier A, Erickson H, Albanil A, Lee M, Kim SW, Bull DA. Polymeric gene delivery of ischemia-inducible VEGF significantly attenuates infarct size and apoptosis following myocardial infarct. Gene therapy. 2009;16:127–135. doi: 10.1038/gt.2008.146. [DOI] [PubMed] [Google Scholar]
  • 128.Schwarz ER, Speakman MT, Patterson M, Hale SS, Isner JM, Kedes LH, Kloner RA. Evaluation of the effects of intramyocardial injection of DNA expressing vascular endothelial growth factor (VEGF) in a myocardial infarction model in the rat--angiogenesis and angioma formation. Journal of the American College of Cardiology. 2000;35:1323–1330. doi: 10.1016/s0735-1097(00)00522-2. [DOI] [PubMed] [Google Scholar]
  • 129.Chang CW, Christensen LV, Lee M, Kim SW. Efficient expression of vascular endothelial growth factor using minicircle DNA for angiogenic gene therapy. Journal of controlled release : official journal of the Controlled Release Society. 2008;125:155–163. doi: 10.1016/j.jconrel.2007.10.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Maiese K, Li F, Chong ZZ. New avenues of exploration for erythropoietin. JAMA : the journal of the American Medical Association. 2005;293:90–95. doi: 10.1001/jama.293.1.90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Hanlon PR, Fu P, Wright GL, Steenbergen C, Arcasoy MO, Murphy E. Mechanisms of erythropoietin-mediated cardioprotection during ischemia-reperfusion injury: role of protein kinase C and phosphatidylinositol 3-kinase signaling. FASEB journal : official publication of the Federation of American Societies for Experimental Biology. 2005;19:1323–1325. doi: 10.1096/fj.04-3545fje. [DOI] [PubMed] [Google Scholar]
  • 132.Lipsic E, Schoemaker RG, van der Meer P, Voors AA, van Veldhuisen DJ, van Gilst WH. Protective effects of erythropoietin in cardiac ischemia: from bench to bedside. Journal of the American College of Cardiology. 2006;48:2161–2167. doi: 10.1016/j.jacc.2006.08.031. [DOI] [PubMed] [Google Scholar]
  • 133.Parsa CJ, Matsumoto A, Kim J, Riel RU, Pascal LS, Walton GB, Thompson RB, Petrofski JA, Annex BH, Stamler JS, Koch WJ. A novel protective effect of erythropoietin in the infarcted heart. The Journal of clinical investigation. 2003;112:999–1007. doi: 10.1172/JCI18200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Richard-Fiardo P, Payen E, Chevre R, Zuber J, Letrou-Bonneval E, Beuzard Y, Pitard B. Therapy of anemia in kidney failure, using plasmid encoding erythropoietin. Human gene therapy. 2008;19:331–342. doi: 10.1089/hum.2006.0101. [DOI] [PubMed] [Google Scholar]
  • 135.Levin NW, Fishbane S, Canedo FV, Zeig S, Nassar GM, Moran JE, Villa G, Beyer U, Oguey D, M.s. investigators Intravenous methoxy polyethylene glycol-epoetin beta for haemoglobin control in patients with chronic kidney disease who are on dialysis: a randomised non-inferiority trial (MAXIMA) Lancet. 2007;370:1415–1421. doi: 10.1016/S0140-6736(07)61599-2. [DOI] [PubMed] [Google Scholar]
  • 136.Elliott S, Pham E, Macdougall IC. Erythropoietins: a common mechanism of action. Experimental hematology. 2008;36:1573–1584. doi: 10.1016/j.exphem.2008.08.003. [DOI] [PubMed] [Google Scholar]
  • 137.Lee Y, Nam HY, Kim J, Lee M, Yockman JW, Shin SK, Kim SW. Human erythropoietin gene delivery using an arginine-grafted bioreducible polymer system. Molecular therapy : the journal of the American Society of Gene Therapy. 2012;20:1360–1366. doi: 10.1038/mt.2012.62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Nam HY, Lee Y, Lee M, Shin SK, Kim TI, Kim SW, Bull DA. Erythropoietin gene delivery using an arginine-grafted bioreducible polymer system. Journal of controlled release : official journal of the Controlled Release Society. 2012;157:437–444. doi: 10.1016/j.jconrel.2011.10.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Namiuchi S, Kagaya Y, Ohta J, Shiba N, Sugi M, Oikawa M, Kunii H, Yamao H, Komatsu N, Yui M, Tada H, Sakuma M, Watanabe J, Ichihara T, Shirato K. High serum erythropoietin level is associated with smaller infarct size in patients with acute myocardial infarction who undergo successful primary percutaneous coronary intervention. Journal of the American College of Cardiology. 2005;45:1406–1412. doi: 10.1016/j.jacc.2005.01.043. [DOI] [PubMed] [Google Scholar]
  • 140.Taniguchi N, Nakamura T, Sawada T, Matsubara K, Furukawa K, Hadase M, Nakahara Y, Nakamura T, Matsubara H. Erythropoietin prevention trial of coronary restenosis and cardiac remodeling after ST-elevated acute myocardial infarction (EPOC-AMI): a pilot, randomized, placebo-controlled study. Circulation journal : official journal of the Japanese Circulation Society. 2010;74:2365–2371. doi: 10.1253/circj.cj-10-0267. [DOI] [PubMed] [Google Scholar]
  • 141.Voors AA, Belonje AM, Zijlstra F, Hillege HL, Anker SD, Slart RH, Tio RA, van 't Hof A, Jukema JW, Peels HO, Henriques JP, Ten Berg JM, Vos J, van Gilst WH, van Veldhuisen DJ, H.I. Investigators A single dose of erythropoietin in ST-elevation myocardial infarction. European heart journal. 2010;31:2593–2600. doi: 10.1093/eurheartj/ehq304. [DOI] [PubMed] [Google Scholar]
  • 142.Lee Y, McGinn AN, Olsen CD, Nam K, Lee M, Shin SK, Kim SW. Human erythropoietin gene delivery for cardiac remodeling of myocardial infarction in rats. Journal of controlled release : official journal of the Controlled Release Society. 2013;171:24–32. doi: 10.1016/j.jconrel.2013.06.022. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES