1
|
Gene Therapy and Cardiovascular Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1396:235-254. [DOI: 10.1007/978-981-19-5642-3_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
|
2
|
Liu J, Sun Q, Wang Y, Cui J, Zhang M, Li L, Jia H, Zhang L, Zhu B, Jiang M, Yu B, Zhang S. ERK1/2 pathway regulates coxsackie and adenovirus receptor expression in mouse cardiac stem cells. Exp Ther Med 2017; 13:3348-3354. [PMID: 28587412 PMCID: PMC5450675 DOI: 10.3892/etm.2017.4414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 10/10/2016] [Indexed: 11/05/2022] Open
Abstract
Cardiac stem cells (CSCs) are the most promising and effective candidates for the therapy of cardiac regenerative diseases; however, they have marked limitations. For instance, the implantation of CSCs is hampered by factors such as their sustainability and long-term durability. Gene modification appears to be the most effective method of optimizing CSCs and gene therapy trials have demonstrated that efficient gene transfer is key to achieving therapeutic efficacy. However, the transduction ability of adenovirus (Ad) is limited. Previous studies have reported that low expression of coxsackie and adenovirus receptor (CAR) in target cells decreases the transduction efficiency. A promising method for improving Ad-mediated gene transfer is to increase CAR expression in target cells. The present study investigated the effect of the Raf-mitogen-associated protein kinase (MAPK) kinase (MEK)-extracellular signal-associated protein kinase (ERK) signaling pathway on the expression of CAR on CSCs, as this pathway decreases cell-cell adhesion via cell surface molecules. The results demonstrated that interference with the Raf-MEK-ERK signaling pathway by knockdown of ERK1/2 upregulated the expression of CAR. The entry of the Ad into the cells was increased following inhibition of ERK1/2. Moreover, following knockdown of CAR, the entry of Ad into cells was decreased. However, knockdown of c-Jun N-terminal kinase and p38 as other components of the MAPK pathway did not affect CAR expression. Therefore, CAR expression in CSCs may be mediated via the Raf-MEK-ERK signaling pathway. Upregulation of CAR by knockdown of ERK1/2 may significantly improve Ad-mediated genetic modification of CSCs in the treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Jingjin Liu
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China.,Key Laboratories of Education, Ministry for Myocardial Ischemia Mechanism and Treatment (Harbin Medical University), Ministry of Education, Harbin, Heilongjiang 150086, P.R. China
| | - Qiang Sun
- Department of Cardiology, The First Hospital of Qinhuangdao, Qinhuangdao, Hebei 066000, P.R. China
| | - Yongshun Wang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China.,Key Laboratories of Education, Ministry for Myocardial Ischemia Mechanism and Treatment (Harbin Medical University), Ministry of Education, Harbin, Heilongjiang 150086, P.R. China
| | - Jinjin Cui
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China.,Key Laboratories of Education, Ministry for Myocardial Ischemia Mechanism and Treatment (Harbin Medical University), Ministry of Education, Harbin, Heilongjiang 150086, P.R. China
| | - Maomao Zhang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China.,Key Laboratories of Education, Ministry for Myocardial Ischemia Mechanism and Treatment (Harbin Medical University), Ministry of Education, Harbin, Heilongjiang 150086, P.R. China
| | - Lili Li
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Haibo Jia
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Lulu Zhang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Bin Zhu
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Miaomiao Jiang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Bo Yu
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China.,Key Laboratories of Education, Ministry for Myocardial Ischemia Mechanism and Treatment (Harbin Medical University), Ministry of Education, Harbin, Heilongjiang 150086, P.R. China
| | - Shuo Zhang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| |
Collapse
|
3
|
Precision Medicine, Cardiovascular Disease and Hunting Elephants. Prog Cardiovasc Dis 2016; 58:651-60. [DOI: 10.1016/j.pcad.2016.02.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 02/17/2016] [Indexed: 01/14/2023]
|
4
|
Abstract
Heart failure is a significant burden to the global healthcare system and represents an underserved market for new pharmacologic strategies, especially therapies which can address root cause myocyte dysfunction. Modern drugs, surgeries, and state-of-the-art interventions are costly and do not improve survival outcome measures. Gene therapy is an attractive strategy, whereby selected gene targets and their associated regulatory mechanisms can be permanently managed therapeutically in a single treatment. This in theory could be sustainable for the patient's life. Despite the promise, however, gene therapy has numerous challenges that must be addressed together as a treatment plan comprising these key elements: myocyte physiologic target validation, gene target manipulation strategy, vector selection for the correct level of manipulation, and carefully utilizing an efficient delivery route that can be implemented in the clinic to efficiently transfer the therapy within safety limits. This chapter summarizes the key developments in cardiac gene therapy from the perspective of understanding each of these components of the treatment plan. The latest pharmacologic gene targets, gene therapy vectors, delivery routes, and strategies are reviewed.
Collapse
Affiliation(s)
- Anthony S Fargnoli
- Icahn School of Medicine at Mount Sinai, Cardiovascular Research Center, New York, NY, USA.
| | - Michael G Katz
- Icahn School of Medicine at Mount Sinai, Cardiovascular Research Center, New York, NY, USA
| | - Charles R Bridges
- Icahn School of Medicine at Mount Sinai, Cardiovascular Research Center, New York, NY, USA
| | - Roger J Hajjar
- Icahn School of Medicine at Mount Sinai, Cardiovascular Research Center, New York, NY, USA
| |
Collapse
|
5
|
Chen T, Zhou G, Zhou Q, Tang H, Ibe JCF, Cheng H, Gou D, Chen J, Yuan JXJ, Raj JU. Loss of microRNA-17∼92 in smooth muscle cells attenuates experimental pulmonary hypertension via induction of PDZ and LIM domain 5. Am J Respir Crit Care Med 2015; 191:678-92. [PMID: 25647182 DOI: 10.1164/rccm.201405-0941oc] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
RATIONALE Recent studies suggest that microRNAs (miRNAs) play important roles in regulation of pulmonary artery smooth muscle cell (PASMC) phenotype and are implicated in pulmonary arterial hypertension (PAH). However, the underlying molecular mechanisms remain elusive. OBJECTIVES This study aims to understand the mechanisms regulating PASMC proliferation and differentiation by microRNA-17∼92 (miR-17∼92) and to elucidate its implication in PAH. METHODS We generated smooth muscle cell (SMC)-specific miR-17∼92 and PDZ and LIM domain 5 (PDLIM5) knockout mice and overexpressed miR-17∼92 and PDLIM5 by injection of miR-17∼92 mimics or PDLIM5-V5-His plasmids and measured their responses to hypoxia. We used miR-17∼92 mimics, inhibitors, overexpression vectors, small interfering RNAs against PDLIM5, Smad, and transforming growth factor (TGF)-β to determine the role of miR-17∼92 and its downstream targets in PASMC proliferation and differentiation. MEASUREMENTS AND MAIN RESULTS We found that human PASMC (HPASMC) from patients with PAH expressed decreased levels of the miR-17∼92 cluster, TGF-β, and SMC markers. Overexpression of miR-17∼92 increased and restored the expression of TGF-β3, Smad3, and SMC markers in HPASMC of normal subjects and patients with idiopathic PAH, respectively. Knockdown of Smad3 but not Smad2 prevented miR-17∼92-induced expression of SMC markers. SMC-specific knockout of miR-17∼92 attenuated hypoxia-induced pulmonary hypertension (PH) in mice, whereas reconstitution of miR-17∼92 restored hypoxia-induced PH in these mice. We also found that PDLIM5 is a direct target of miR-17/20a, and hypertensive HPASMC and mouse PASMC expressed elevated PDLIM5 levels. Suppression of PDLIM5 increased expression of SMC markers and enhanced TGF-β/Smad2/3 activity in vitro and enhanced hypoxia-induced PH in vivo, whereas overexpression of PDLIM5 attenuated hypoxia-induced PH. CONCLUSIONS We provided the first evidence that miR-17∼92 inhibits PDLIM5 to induce the TGF-β3/SMAD3 pathway, contributing to the pathogenesis of PAH.
Collapse
|
6
|
Scimia MC, Cannavo A, Koch WJ. Gene therapy for heart disease: molecular targets, vectors and modes of delivery to myocardium. Expert Rev Cardiovasc Ther 2014; 11:999-1013. [PMID: 23984926 DOI: 10.1586/14779072.2013.818813] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Despite the numerous hurdles that gene therapy has encountered along the way, clinical trials over the last few years are showing promising results in many fields of medicine, including cardiology, where many targets are moving toward clinical development. In this review, the authors discuss the current state of the art in terms of clinical and preclinical development. They also examine vector technology and available vector-delivery strategies.
Collapse
Affiliation(s)
- Maria Cecilia Scimia
- Department of Pharmacology, Center for Translational Medicine, Temple University School of Medicine, 3500 N Broad St, MERB 941, Philadelphia, PA 19140, USA
| | | | | |
Collapse
|
7
|
Liu M, Li M, Sun S, Li B, Du D, Sun J, Cao F, Li H, Jia F, Wang T, Chang N, Yu H, Wang Q, Peng H. The use of antibody modified liposomes loaded with AMO-1 to deliver oligonucleotides to ischemic myocardium for arrhythmia therapy. Biomaterials 2014; 35:3697-707. [PMID: 24468403 DOI: 10.1016/j.biomaterials.2013.12.099] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2013] [Accepted: 12/28/2013] [Indexed: 01/07/2023]
Abstract
MicroRNA-1 (miR-1) has been found in cardiac and skeletal tissues. It is overexpressed in ischemic cardiac tissues. Down-regulation of miR-1 could relieve arrhythmogenesis by the anti-miR-1 antisense oligonucleotides (AMO-1). To increase the therapeutic efficiency and inhibit off-target effects of AMO-1, here we explored anti-cardiac troponin I (cTnI) antibody modified liposomes loading with AMO-1 (cT-A-LIP) to deliver the oligonucleotides to ischemic myocardium tissues. Liposomal cytotoxicity was assessed by MTT assay. The targeting abilities to foci were evaluated by in vivo imaging. The uptake and bio-distribution in vitro were observed by live cell station and flow cytometry, respectively. The anti-arrhythmic effects of cT-A-LIP in vivo were evaluated by electrocardiograms (ECG), immunohistochemistry, real-time PCR and patch-clamp recording. Immunohistochemistry showed that cTnI expression had a peak at the third day after myocardial infarction (MI). After cT-LIP administration via tail vein, accumulation of fluorescent trackers in the ischemic foci was significantly increased more than that of LIP. In addition, after cT-A-LIP administration, the ischemic arrhythmias were recovered and ST segment in ECG was elevated nearly back to normal. Compared with MI group, miR-1 expression was significantly down-regulated while Kir2.1 and CX43 protein expression were increased. Patch-clamp recordings showed that cT-A-LIP as well as AMO-1 incubation increased K(+) current density in guinea pigs ventricular cardiomyocytes acting on repolarized membrane potential. In conclusion, the cT-A-LIP not only delivered AMO-1 to ischemic myocardium in MI rats, but validated AMO-1 on relieving ischemic arrhythmia by silencing of miR-1 in ischemic myocardium and restoring the depolarized resting membrane potential (RMP) in MI rats.
Collapse
Affiliation(s)
- Meifang Liu
- Department of Pharmaceutics, Daqing Campus of Harbin Medical University, Daqing 163319, China
| | - Minghui Li
- Department of Pharmaceutics, Daqing Campus of Harbin Medical University, Daqing 163319, China
| | - Shouli Sun
- Department of Pharmaceutics, Daqing Campus of Harbin Medical University, Daqing 163319, China
| | - Baoxin Li
- Department of Pharmaceutics, Harbin Medical University, Harbin 150086, China
| | - Dan Du
- Department of Pharmaceutics, Daqing Campus of Harbin Medical University, Daqing 163319, China
| | - Jialiang Sun
- Department of Pharmaceutics, Harbin Medical University, Harbin 150086, China
| | - Fangyuan Cao
- Department of Pharmaceutics, Daqing Campus of Harbin Medical University, Daqing 163319, China
| | - Haichun Li
- Department of Pharmaceutics, Daqing Campus of Harbin Medical University, Daqing 163319, China
| | - Feng Jia
- Department of Chemical and Biological Engineering, Ames, IA 50011, USA
| | - Tianfu Wang
- Department of Chemical and Biological Engineering, Ames, IA 50011, USA
| | - Naidan Chang
- Department of Pharmaceutics, Daqing Campus of Harbin Medical University, Daqing 163319, China
| | - Hui Yu
- Department of Pharmaceutics, Daqing Campus of Harbin Medical University, Daqing 163319, China
| | - Qun Wang
- Department of Chemical and Biological Engineering, Ames, IA 50011, USA; Department of Civil, Construction and Environmental Engineering, Iowa State University, Ames, IA 50011, USA
| | - Haisheng Peng
- Department of Pharmaceutics, Daqing Campus of Harbin Medical University, Daqing 163319, China; Department of Chemical and Biological Engineering, Ames, IA 50011, USA.
| |
Collapse
|
8
|
Chiu B, Li B, Chow TWS. Novel 3D ultrasound image-based biomarkers based on a feature selection from a 2D standardized vessel wall thickness map: a tool for sensitive assessment of therapies for carotid atherosclerosis. Phys Med Biol 2013; 58:5959-82. [DOI: 10.1088/0031-9155/58/17/5959] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
9
|
Wen Q, Zhou L, Zhou C, Zhou M, Luo W, Ma L. Change in hepatocyte growth factor concentration promote mesenchymal stem cell-mediated osteogenic regeneration. J Cell Mol Med 2012; 16:1260-73. [PMID: 21831134 PMCID: PMC3823079 DOI: 10.1111/j.1582-4934.2011.01407.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Mesenchymal stem cells (MSCs) play a crucial role in tissue repair by secretion of tissue nutrient factors such as hepatocyte growth factor (HGF). However, studies examining the effects of HGF on the proliferation and differentiation of MSCs used different concentrations of HGF and reported conflicting conclusions. This study aimed to determine the mechanisms by which different concentrations of HGF regulate MSC proliferation and osteogenic differentiation, and validate the mechanism in an animal model of early stage avascular necrosis of femoral head (ANFH). Our results demonstrate that a low concentration of HGF (20 ng/ml) preferentially promotes MSC osteogenic differentiation through increased c-Met expression and phosphorylation, Akt pathway activation, and increased expression of p27, Runx2 and Osterix. In contrast, a high concentration of HGF (100 ng/ml) strongly induced proliferation by inducing strong activation of the ERK1/2 signalling pathway. As validated by animal experiments, high localized expression of HGF achieved by transplantation of HGF transgenic MSCs into ANFH rabbits increased the number of MSCs. Subsequently, 2 weeks after transplantation, HGF levels decreased and MSCs differentiated into osteoblasts and resulted in efficient tissue repair. Our results demonstrate that sequential concentration changes in HGF control the proliferation and osteogenic differentiation of MSCs in vivo. This phenomenon can be exploited therapeutically to induce bone regeneration and, in turn, improve the efficacy of pharmacological intervention for ANFH treatment.
Collapse
Affiliation(s)
- Qian Wen
- Institute of Molecular Immunology, School of Biotechnology, Southern Medical University, Guangzhou, China.
| | | | | | | | | | | |
Collapse
|
10
|
Sun J, Li X, Feng H, Gu H, Blair T, Li J, Soriano S, Meng Y, Zhang F, Feng Q, Yang X. Magnetic resonance imaging of bone marrow cell-mediated interleukin-10 gene therapy of atherosclerosis. PLoS One 2011; 6:e24529. [PMID: 21915349 PMCID: PMC3168522 DOI: 10.1371/journal.pone.0024529] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2011] [Accepted: 08/12/2011] [Indexed: 11/23/2022] Open
Abstract
Background A characteristic feature of atherosclerosis is its diffuse involvement of arteries across the entire human body. Bone marrow cells (BMC) can be simultaneously transferred with therapeutic genes and magnetic resonance (MR) contrast agents prior to their transplantation. Via systemic transplantation, these dual-transferred BMCs can circulate through the entire body and thus function as vehicles to carry genes/contrast agents to multiple atherosclerosis. This study was to evaluate the feasibility of using in vivo MR imaging (MRI) to monitor BMC-mediated interleukin-10 (IL-10) gene therapy of atherosclerosis. Methodology For in vitro confirmation, donor mouse BMCs were transduced by IL-10/lentivirus, and then labeled with a T2-MR contrast agent (Feridex). For in vivo validation, atherosclerotic apoE−/− mice were intravenously transplanted with IL-10/Feridex-BMCs (Group I, n = 5) and Feridex-BMCs (Group II, n = 5), compared to controls without BMC transplantation (Group III, n = 5). The cell migration to aortic atherosclerotic lesions was monitored in vivo using 3.0T MRI with subsequent histology correlation. To evaluate the therapeutic effect of BMC-mediated IL-10 gene therapy, we statistically compared the normalized wall indexes (NWI) of ascending aortas amongst different mouse groups with various treatments. Principal Findings Of in vitro experiments, simultaneous IL-10 transduction and Feridex labeling of BMCs were successfully achieved, with high cell viability and cell labeling efficiency, as well as IL-10 expression efficiency (≥90%). Of in vivo experiments, MRI of animal groups I and II showed signal voids within the aortic walls due to Feridex-created artifacts from the migrated BMCs in the atherosclerotic plaques, which were confirmed by histology. Histological quantification showed that the mean NWI of group I was significantly lower than those of group II and group III (P<0.05). Conclusion This study has confirmed the possibility of using MRI to track, in vivo, IL-10/Feridex-BMCs recruited to atherosclerotic lesions, where IL-10 genes function to prevent the progression of atherosclerosis.
Collapse
Affiliation(s)
- Jihong Sun
- Department of Radiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Image-Guided Bio-Molecular Interventions Section, Department of Radiology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Xubin Li
- Image-Guided Bio-Molecular Interventions Section, Department of Radiology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Hongqing Feng
- Image-Guided Bio-Molecular Interventions Section, Department of Radiology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Huidong Gu
- Image-Guided Bio-Molecular Interventions Section, Department of Radiology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Tiffany Blair
- Image-Guided Bio-Molecular Interventions Section, Department of Radiology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Jiakai Li
- Image-Guided Bio-Molecular Interventions Section, Department of Radiology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Stephanie Soriano
- Image-Guided Bio-Molecular Interventions Section, Department of Radiology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Yanfeng Meng
- Image-Guided Bio-Molecular Interventions Section, Department of Radiology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Feng Zhang
- Image-Guided Bio-Molecular Interventions Section, Department of Radiology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Qinghua Feng
- Department of Pathology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Xiaoming Yang
- Image-Guided Bio-Molecular Interventions Section, Department of Radiology, University of Washington School of Medicine, Seattle, Washington, United States of America
- * E-mail:
| |
Collapse
|
11
|
Gupta AS. Nanomedicine approaches in vascular disease: a review. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2011; 7:763-79. [PMID: 21601009 DOI: 10.1016/j.nano.2011.04.001] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2011] [Revised: 03/11/2011] [Accepted: 04/05/2011] [Indexed: 01/26/2023]
Abstract
UNLABELLED Nanomedicine approaches have revolutionized the treatment of cancer and vascular diseases, where the limitations of rapid nonspecific clearance, poor biodistribution and harmful side effects associated with direct systemic drug administration can be overcome by packaging the agents within sterically stabilized, long-circulating nanovehicles that can be further surface-modified with ligands to actively target cellular/molecular components of the disease. With significant advancements in genetics, proteomics, cellular and molecular biology and biomaterials engineering, the nanomedicine strategies have become progressively refined regarding the modulation of surface and bulk chemistry of the nanovehicles, control of drug release kinetics, manipulation of nanoconstruct geometry and integration of multiple functionalities on single nanoplatforms. The current review aims to capture the various nanomedicine approaches directed specifically toward vascular diseases during the past two decades. Analysis of the promises and limitations of these approaches will help identify and optimize vascular nanomedicine systems to enhance their efficacy and clinical translation in the future. FROM THE CLINICAL EDITOR Nanomedicine-based approaches have had a major impact on the treatment and diagnosis of malignancies and vascular diseases. This review discusses various nanomedicine approaches directed specifically toward vascular diseases during the past two decades, highlighting their advantages, limitations and offering new perspectives on future applications.
Collapse
Affiliation(s)
- Anirban Sen Gupta
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio 44106, USA.
| |
Collapse
|
12
|
Tlaxca JL, Anderson CR, Klibanov AL, Lowrey B, Hossack JA, Alexander JS, Lawrence MB, Rychak JJ. Analysis of in vitro transfection by sonoporation using cationic and neutral microbubbles. ULTRASOUND IN MEDICINE & BIOLOGY 2010; 36:1907-1918. [PMID: 20800945 PMCID: PMC2996233 DOI: 10.1016/j.ultrasmedbio.2010.05.014] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2010] [Revised: 05/13/2010] [Accepted: 05/14/2010] [Indexed: 05/26/2023]
Abstract
The objective of the study was to examine the role of acoustic power intensity and microbubble and plasmid concentrations on transfection efficiency in HEK-293 cells using a sonoporator with a 1-MHz transducer. A green fluorescent protein (GFP) reporter plasmid was delivered in as much as 80% of treated cells, and expression of the GFP protein was observed in as much as 75% of cells, using a power intensity of 2 W/cm(2) with a 25% duty cycle. In addition, the relative transfection abilities of a lipid noncationic and cationic microbubble platform were investigated. As a positive control, cells were transfected using Lipofectamine reagent. Cell survival and transfection efficiency were inversely proportional to acoustic power and microbubble concentration. Our results further demonstrated that high-efficiency transfection could be achieved, but at the expense of cell loss. Moreover, direct conjugation of plasmid to the microbubble did not appear to significantly enhance transfection efficiency under the examined conditions, although this strategy may be important for targeted transfection in vivo.
Collapse
Affiliation(s)
- Jose L. Tlaxca
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA
| | | | - Alexander L. Klibanov
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA
- Cardiovascular Research Center, University of Virginia, Charlottesville, VA
- Cardiovascular Division, Department of Internal medicine, University of Virginia, Charlottesville, VA
| | - Bryce Lowrey
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA
| | - John A. Hossack
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA
| | - J. Steven Alexander
- Department of Molecular and Cellular Physiology, Louisiana State University Health Science center, Shreveport, LA
| | - Michael B. Lawrence
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA
| | | |
Collapse
|
13
|
Dual transfer of GFP gene and MGd into stem-progenitor cells: toward in vivo MRI of stem cell-mediated gene therapy of atherosclerosis. Acad Radiol 2010; 17:547-52. [PMID: 20227305 DOI: 10.1016/j.acra.2010.02.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2009] [Revised: 02/05/2010] [Accepted: 02/07/2010] [Indexed: 01/08/2023]
Abstract
RATIONALE AND OBJECTIVES The aim of this study was to develop a new technique, the use of magnetic resonance (MR) imaging (MRI) to monitor gene/MR-cotransferred stem-progenitor cells (SPCs) recruited to atherosclerosis. MATERIALS AND METHODS First, a green fluorescent protein (GFP) gene and a T1 MR contrast agent (motexafin gadolinium [MGd]) were cotransferred into neural or bone marrow (BM)-derived SPCs. GFP expression and MGd signal were confirmed by fluorescent microscopy and quantified by flow cytometry. Cell viability and proliferation were then evaluated by trypan blue exclusion and 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium assay, and GFP/MGd-transferred cells were imaged using 1.5-T and 9.4-T MR scanners. For in vivo validation, GFP/MGd-cotransferred beta-galactosidase-BM SPCs were transplanted to apolipoprotein E-knockout mice, and cell migration to atherosclerotic aortas was monitored using 9.4-T micro-MRI with subsequent histologic correlations. RESULTS Fluorescent microscopy demonstrated simultaneous GFP expression and MGd signals in cotransferred-cells. Quantitative flow cytometry showed GFP-positive cells at 47 +/- 25% and 56 +/- 12% and MGd-positive cells at 96 +/- 6% and 57 +/- 11% for neural stem cells and BM cells, respectively. Cell viability and metabolic rates of cotransferred cells were 86 +/- 4% and 84 +/- 12%, respectively. In vivo MRI revealed high MR signals of the aortic walls in GFP/MGd-transferred mice, which were confirmed by histologic correlations. CONCLUSION This study has initially proven the new concept of MRI for plaque-specific, cell-mediated gene expression of atherosclerosis.
Collapse
|
14
|
Ni J, Waldman A, Khachigian LM. c-Jun regulates shear- and injury-inducible Egr-1 expression, vein graft stenosis after autologous end-to-side transplantation in rabbits, and intimal hyperplasia in human saphenous veins. J Biol Chem 2009; 285:4038-4048. [PMID: 19940138 DOI: 10.1074/jbc.m109.078345] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Coronary artery bypass graft failure represents an unsolved problem in interventional cardiology and heart surgery. Late occlusion of autologous saphenous vein bypass grafts is a consequence of neointima formation underpinned by smooth muscle cell (SMC) migration and proliferation. Poor long term patency and the lack of pharmacologic agents that prevent graft failure necessitate effective alternative therapies. Our objective here was to evaluate the effect of targeted inhibition of the bZIP transcription factor c-Jun on intimal hyperplasia in human saphenous veins and vein graft stenosis after autologous end-to-side transplantation. DNAzymes targeting c-Jun attenuated intimal hyperplasia in human saphenous vein explants. Adenovirus-forced c-Jun expression stimulated SMC proliferation, proliferating cell nuclear antigen, and MMP-2 expression. c-Jun DNAzymes abrogated Adeno-c-Jun-inducible SMC growth and wound repair and reduced intimal thickening in jugular veins of New Zealand white rabbits 4 weeks after autologous end-to-side transplantation to carotid arteries. Conversely, in a DNAzyme-free setting, Adeno-c-Jun potentiated neointima formation in the veins compared with Adeno-LacZ. Inducible c-Jun expression is ERK1/2- and JNK-dependent but p38-independent. Injury- and shear-inducible c-Jun controls early growth response-1. These data demonstrate that strategies targeting c-Jun may be useful for the prevention of vein graft stenosis. Control of one important shear-responsive transcription factor by another indicates the existence of transcriptional amplification mechanisms that magnify the vascular response to cell injury or stress through inducible transcriptional networks.
Collapse
Affiliation(s)
- Jun Ni
- From the Centre for Vascular Research, University of New South Wales, Sydney NSW 2052, Australia
| | - Alla Waldman
- From the Centre for Vascular Research, University of New South Wales, Sydney NSW 2052, Australia
| | - Levon M Khachigian
- From the Centre for Vascular Research, University of New South Wales, Sydney NSW 2052, Australia.
| |
Collapse
|
15
|
Boutin V, Legrand A, Mayer R, Nachtigal M, Monsigny M, Midoux P. Glycofection: The Ubiquitous Roles of Sugar Bound on Glycoplexes. Drug Deliv 2008. [DOI: 10.1080/107175499267156] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022] Open
|
16
|
Ly HQ, Kawase Y, Hajjar RJ. Advances in gene-based therapy for heart failure. J Cardiovasc Transl Res 2008; 1:127-36. [PMID: 20559907 DOI: 10.1007/s12265-008-9022-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2008] [Accepted: 03/17/2008] [Indexed: 01/08/2023]
Abstract
Heart failure is a major cause of morbidity and mortality in western countries. While progress in current treatment modalities is making steady and incremental gains to reduce this disease burden, there remains a need to explore novel therapeutic strategies. Clinicians and researchers alike have thus looked towards novel adjunctive therapeutic strategies, including gene-based therapy for congestive heart failure (CHF). Advances in the understanding of the molecular basis of CHF, combined to the evolution of increasingly efficient gene transfer technology, have placed congestive heart failure within reach of gene-based therapy. This review will discuss issues related to gene vector systems, gene delivery strategies, and gene targets for intervention in the setting of CHF.
Collapse
Affiliation(s)
- Hung Q Ly
- Cardiovascular Research Center, Mount Sinai School of Medicine, One Gustave L. Levy Place, P.O. Box 1030, New York, NY 10029, USA
| | | | | |
Collapse
|
17
|
Kalka C, Baumgartner I. Gene and stem cell therapy in peripheral arterial occlusive disease. Vasc Med 2008; 13:157-72. [DOI: 10.1177/1358863x08088616] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Abstract Peripheral arterial occlusive disease (PAOD) is a manifestation of systemic atherosclerosis strongly associated with a high risk of cardiovascular morbidity and mortality. In a considerable proportion of patients with PAOD, revascularization either by endovascular means or by open surgery combined with best possible risk factor modification does not achieve limb salvage or relief of ischaemic rest pain. As a consequence, novel therapeutic strategies have been developed over the last two decades aiming to promote neovascularization and remodelling of collaterals. Gene and stem cell therapy are the main directions for clinical investigation concepts. For both, preclinical studies have shown promising results using a wide variety of genes encoding for growth factors and populations of adult stem cells, respectively. As a consequence, clinical trials have been performed applying gene and stem cell-based concepts. However, it has become apparent that a straightforward translation into humans is not possible. While several trials reported relief of symptoms and functional improvement, other trials did not confirm this early promise of efficacy. Ongoing clinical trials with an improved study design are needed to confirm the potential that gene and cell therapy may have and to prevent the gaps in our scientific knowledge that will jeopardize the establishment of angiogenic therapy as an additional medical treatment of PAOD. This review summarizes the experimental background and presents the current status of clinical applications and future perspectives of the therapeutic use of gene and cell therapy strategies for PAOD.
Collapse
Affiliation(s)
- C Kalka
- Division of Vascular Medicine, Swiss Cardiovascular Center, University Hospital of Bern, Switzerland
| | - Iris Baumgartner
- Division of Vascular Medicine, Swiss Cardiovascular Center, University Hospital of Bern, Switzerland
| |
Collapse
|
18
|
Haider HK, Elmadbouh I, Jean-Baptiste M, Ashraf M. Nonviral vector gene modification of stem cells for myocardial repair. Mol Med 2008; 14:79-86. [PMID: 17985003 DOI: 10.2119/2007-00092.haider] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2007] [Accepted: 10/19/2007] [Indexed: 01/09/2023] Open
Abstract
Therapeutic angiogenesis and myogenesis restore perfusion of ischemic myocardium and improve left ventricular contractility. These therapeutic modalities must be considered as complementary rather than competing to exploit their advantages for optimal beneficial effects. The resistant nature of cardiomyocytes to gene transfection can be overcome by ex vivo delivery of therapeutic genes to the heart using genetically modified stem cells. This review article gives an overview of different vectors and delivery systems in general used for therapeutic gene delivery to the heart and provides a critical appreciation of the ex vivo gene delivery approach using genetically modified stem cells to achieve angiomyogenesis for the treatment of infarcted heart.
Collapse
Affiliation(s)
- Husnain K Haider
- Department of Pathology and Laboratory Medicine, University of Cincinnati, OH 45267-0529, USA
| | | | | | | |
Collapse
|
19
|
Affiliation(s)
- David M. Kaye
- Heart Failure Research Group, Baker Heart Research Institute, Melbourne, Victoria 8008, Australia;
| | - Masahiko Hoshijima
- Institute of Molecular Medicine, University of California, San Diego, La Jolla, California 92093-0346
| | - Kenneth R. Chien
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Stem Cell Institute, Harvard Medical School, Richard B. Simches Research Centre, Boston, Massachusetts 02114;
| |
Collapse
|
20
|
Muto A, Fitzgerald TN, Pimiento JM, Maloney S, Teso D, Paszkowiak JJ, Westvik TS, Kudo FA, Nishibe T, Dardik A. Smooth muscle cell signal transduction: implications of vascular biology for vascular surgeons. J Vasc Surg 2007; 45 Suppl A:A15-24. [PMID: 17544020 PMCID: PMC1939976 DOI: 10.1016/j.jvs.2007.02.061] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2007] [Accepted: 02/17/2007] [Indexed: 12/31/2022]
Abstract
Vascular smooth muscle cells exhibit varied responses after vessel injury and surgical interventions, including phenotypic switching, migration, proliferation, protein synthesis, and apoptosis. Although the source of the smooth muscle cells that accumulate in the vascular wall is controversial, possibly reflecting migration from the adventitia, from the circulating blood, or in situ differentiation, the intracellular signal transduction pathways that control these processes are being defined. Some of these pathways include the Ras-mitogen-activated protein kinase, phosphatidylinositol 3-kinase-Akt, Rho, death receptor-caspase, and nitric oxide pathways. Signal transduction pathways provide amplification, redundancy, and control points within the cell and culminate in biologic responses. We review some of the signaling pathways activated within smooth muscle cells that contribute to smooth muscle cell heterogeneity and development of pathology such as restenosis and neointimal hyperplasia.
Collapse
MESH Headings
- Animals
- Apoptosis
- Bone Marrow Cells/metabolism
- Cell Differentiation
- Cell Movement
- Cell Proliferation
- Constriction, Pathologic/metabolism
- Constriction, Pathologic/pathology
- Extracellular Matrix/metabolism
- Humans
- Hyperplasia/metabolism
- Hyperplasia/pathology
- Muscle, Smooth, Vascular/injuries
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/physiopathology
- Muscle, Smooth, Vascular/surgery
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Phenotype
- Protein Kinases/metabolism
- Signal Transduction
- Stem Cells/metabolism
- Vascular Surgical Procedures/adverse effects
Collapse
Affiliation(s)
- Akihito Muto
- Department of Surgery, Yale University School of Medicine, New Haven, CT, USA
- Department of Interdepartmental Program in Vascular Biology and Transplantation, Yale University School of Medicine, New Haven, CT, USA
| | - Tamara N Fitzgerald
- Department of Surgery, Yale University School of Medicine, New Haven, CT, USA
- Department of Interdepartmental Program in Vascular Biology and Transplantation, Yale University School of Medicine, New Haven, CT, USA
| | - Jose M Pimiento
- Department of Surgery, Yale University School of Medicine, New Haven, CT, USA
- Department of Interdepartmental Program in Vascular Biology and Transplantation, Yale University School of Medicine, New Haven, CT, USA
- Saint Mary’s Hospital, Waterbury, CT, USA
| | - Stephen Maloney
- Department of Surgery, Yale University School of Medicine, New Haven, CT, USA
- Department of Interdepartmental Program in Vascular Biology and Transplantation, Yale University School of Medicine, New Haven, CT, USA
- Saint Mary’s Hospital, Waterbury, CT, USA
| | - Desarom Teso
- Department of Surgery, Yale University School of Medicine, New Haven, CT, USA
- Saint Mary’s Hospital, Waterbury, CT, USA
| | - Jacek J Paszkowiak
- Department of Surgery, Yale University School of Medicine, New Haven, CT, USA
- Saint Mary’s Hospital, Waterbury, CT, USA
| | - Tormod S Westvik
- Department of Surgery, Yale University School of Medicine, New Haven, CT, USA
- Department of Interdepartmental Program in Vascular Biology and Transplantation, Yale University School of Medicine, New Haven, CT, USA
| | - Fabio A Kudo
- Department of Surgery, Yale University School of Medicine, New Haven, CT, USA
- Department of Interdepartmental Program in Vascular Biology and Transplantation, Yale University School of Medicine, New Haven, CT, USA
| | | | - Alan Dardik
- Department of Surgery, Yale University School of Medicine, New Haven, CT, USA
- Department of Interdepartmental Program in Vascular Biology and Transplantation, Yale University School of Medicine, New Haven, CT, USA
- VA Connecticut Healthcare System, West Haven, CT, USA
| |
Collapse
|
21
|
Abstract
Heart failure is a major cause of morbidity and mortality in contemporary societies. Although progress in conventional treatment modalities is making steady and incremental gains to reduce this disease burden, there remains a need to explore new and potentially therapeutic approaches. Gene therapy, for example, was initially envisioned as a treatment strategy for inherited monogenic disorders. It is now apparent that gene therapy has broader potential that also includes acquired polygenic diseases, such as heart failure. Advances in the understanding of the molecular basis of conditions such as these, together with the evolution of increasingly efficient gene transfer technology, has placed congestive heart failure within reach of gene-based therapy.
Collapse
Affiliation(s)
- Hung Ly
- Cardiology Division, Cardiovascular Research Center, the Cardiology Laboratory of Integrative Physiology, Imaging at Massachusetts General Hospital, Boston, MA, USA
| | | | | | | |
Collapse
|
22
|
|
23
|
Gene Therapy. CARDIOVASCULAR MEDICINE 2007. [DOI: 10.1007/978-1-84628-715-2_133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
24
|
Abstract
Since its inception in the 1960s, coronary artery bypass graft (CABG) evolved as one of the most common, best documented, and most effective of all major surgical treatments for ischemic heart disease. Despite its widespread use, however, the outcome is not always completely satisfactory. The objective of this review is to highlight the physical determinants of biomechanical design of CABG so that future procedures would have prolonged patency and better outcome. Our central axiom postulates the existence of a mechanical homeostatic state of the blood vessel, i.e., the variation in vessel wall stresses and strains are relatively small under physiological conditions. Any perturbation of mechanical homeostasis leads to growth and remodeling. In this sense, stenosis and failure of a graft may be viewed as an adaptation process gone awry. We outline the principles of engineering design and discuss the biofluid and biosolid mechanics principles that may have the greatest bearing on mechanical homeostasis and the long-term outcome of CABG.
Collapse
Affiliation(s)
- Ghassan S Kassab
- Department of Biomedical Engineering, University of California, Irvine, California 92697-2715, USA.
| | | |
Collapse
|
25
|
Kar S, Kumar A, Gao F, Qiu B, Zhan X, Yang X. Percutaneous optical imaging system to track reporter gene expression from vasculatures in vivo. JOURNAL OF BIOMEDICAL OPTICS 2006; 11:34008. [PMID: 16822058 PMCID: PMC1501088 DOI: 10.1117/1.2209559] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
This study develops a percutaneous optical imaging system for tracking fluorescent reporter gene expression in vasculatures. We build a percutaneous optical imaging system that primarily comprised a 1.5-mm, semi-rigid, two-port optical probe. The performance of the optical probe is first tested in vitro with cell phantoms, and then the feasibility of the percutaneous optical imaging system is validated in vivo in eight femoral artery segments of two pigs. The green fluorescent protein (GFP) gene is locally delivered into four arterial segments, while saline is delivered to the four contralateral arterial segments as controls. The targeted arteries are localized using color Doppler, and thereafter the optical probe is positioned to the target arterial segments under ultrasound guidance. Optical imaging captures are obtained using different exposure times from 10 to 60 s. Subsequently, the GFP- and saline-targeted arteries are harvested for fluorescent microscopy confirmation. The percutaneous optical probe is successfully positioned at a distance approximately 2 mm from the targets in all eight arteries. The in-vivo imaging shows higher average signal intensity in GFP-treated arteries than in saline-treated arteries. This study demonstrates the potential using the percutaneous optical imaging system to monitor, in vivo, reporter gene expression from vasculatures.
Collapse
Affiliation(s)
- S. Kar
- The Russell H. Morgan Department of Radiology and Radiological Science
- Departments of Biomedical Engineering and
| | - A. Kumar
- The Russell H. Morgan Department of Radiology and Radiological Science
| | - F. Gao
- The Russell H. Morgan Department of Radiology and Radiological Science
| | - B. Qiu
- The Russell H. Morgan Department of Radiology and Radiological Science
| | - X. Zhan
- Gynecology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - X. Yang
- The Russell H. Morgan Department of Radiology and Radiological Science
- Corresponding Author: Xiaoming Yang, MD, PhD, Traylor Building, Room 330, 720 Rutland Avenue, Baltimore, MD 21205, Phone number: (410) 502-6960, FAX number: (443) 287-6730, E-mail:
| |
Collapse
|
26
|
Yan J, Tang GL, Wang R, Messina LM. Optimization of adenovirus-mediated endothelial nitric oxide synthase delivery in rat hindlimb ischemia. Gene Ther 2006; 12:1640-50. [PMID: 16107865 DOI: 10.1038/sj.gt.3302563] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Adenovirus-mediated overexpression of endothelial nitric oxide synthase (eNOS) induces collateral artery development and substantially increases blood flow after induction of experimental acute hindlimb ischemia. However, the optimal technique of gene delivery for this or any other form of gene therapy in limb ischemia is still unknown. The purpose of this study was to determine the effect of the two most commonly used techniques, intra-arterial and intramuscular injection, on blood flow recovery, collateral artery development, and preservation of muscle mass. We compared intra-arterial injection under vascular isolation, intra-arterial injection under transient vascular occlusion, and intramuscular injection of phosphate buffered saline (PBS) or adenovirus encoding either the eNOS (AdeNOS) or LacZ (AdlacZ) gene after induction of acute hindlimb ischemia. Delivery of AdeNOS by both intra-arterial injection techniques increased eNOS activity (22.30 versus 10.56, P<0.01), blood flow (0.90+/-0.02 versus 0.69+/-0.07, P<0.001) and collateral artery development (17.56484 versus 13.74259, P<0.05) more than by intramuscular delivery. Intra-arterial injection under transient vascular occlusion led to better preservation of muscle mass, muscle architecture, and clinical ischemic index, but led to greater transgene expression in distant organs and contralateral limb muscles. Intra-arterial injection of AdeNOS under transient vascular occlusion is the optimal technique to reverse severe hindlimb ischemia in the rat. This is the first systematic comparison of different delivery techniques used in gene therapy of experimental hindlimb ischemia.
Collapse
MESH Headings
- Adenoviridae/genetics
- Animals
- Collateral Circulation
- Endothelium, Vascular/enzymology
- Endothelium, Vascular/virology
- Genetic Engineering
- Genetic Therapy/methods
- Genetic Vectors/administration & dosage
- Genetic Vectors/genetics
- Hindlimb/blood supply
- Hindlimb/diagnostic imaging
- Immunohistochemistry/methods
- Injections, Intra-Arterial
- Injections, Intramuscular
- Ischemia/diagnostic imaging
- Ischemia/enzymology
- Ischemia/therapy
- Male
- Microscopy, Confocal
- Models, Animal
- Muscle, Skeletal/diagnostic imaging
- Muscle, Skeletal/enzymology
- Muscle, Skeletal/virology
- Nitric Oxide Synthase Type III/analysis
- Nitric Oxide Synthase Type III/genetics
- Nitric Oxide Synthase Type III/metabolism
- Radiography
- Random Allocation
- Rats
- Rats, Sprague-Dawley
- Regional Blood Flow
- Reverse Transcriptase Polymerase Chain Reaction
Collapse
Affiliation(s)
- J Yan
- Pacific Vascular Research Laboratory, Department of Surgery, Division of Vascular Surgery, University of California San Francisco, San Francisco, CA 94143, USA
| | | | | | | |
Collapse
|
27
|
Jain JP, Modi S, Domb AJ, Kumar N. Role of polyanhydrides as localized drug carriers. J Control Release 2005; 103:541-63. [PMID: 15820403 DOI: 10.1016/j.jconrel.2004.12.021] [Citation(s) in RCA: 104] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2004] [Accepted: 12/13/2004] [Indexed: 10/25/2022]
Abstract
Many drugs that are administered in an unmodified form by conventional systemic routes fail to reach target organs in an effective concentration, or are not effective over a length of time due to a facile metabolism. Various types of targeting delivery systems and devices have been tried over a long period of time to overcome these problems. Targeted delivery or localized drug delivery offers an advantage of reduced body burden and systemic toxicity of the drugs, especially useful for highly toxic drugs like anticancer agents. Local drug delivery via polymer is a simple approach and hypothesized to avoid the above stated problems. Polyanhydrides are a unique class of polymer for drug delivery because some of them demonstrate a near zero order drug release and relatively rapid biodegradation in vivo. Further, the release rate of polyanhydride fabricated device can be altered over a thousand fold by simple changes in the polymer backbone. Hence, these are one of the best-suited polymers for drug delivery, with biodegradability and biocompatibility. The review focuses on the advantages of polyanhydride carriers in localized drug delivery along with their degradability behavior, toxicological profile and role in various disease conditions.
Collapse
Affiliation(s)
- Jay Prakash Jain
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Sec. 67, SAS Nagar (Mohalali) Punjab-160062, India
| | | | | | | |
Collapse
|
28
|
|
29
|
Rajagopalan S, Olin JW, Young S, Erikson M, Grossman PM, Mendelsohn FO, Regensteiner JG, Hiatt WR, Annex BH. Design of the Del-1 for Therapeutic Angiogenesis Trial (DELTA-1), a Phase II Multicenter, Double-Blind, Placebo-Controlled Trial of VLTS-589 in Subjects with Intermittent Claudication Secondary to Peripheral Arterial Disease. Hum Gene Ther 2004; 15:619-24. [PMID: 15212720 DOI: 10.1089/104303404323142060] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The objective of this phase II investigation is to assess the safety and efficacy of a plasmid mediated approach to induce angiogenesis/arteriogenesis with the angiomatrix protein Del-1 (developmentally regulated endothelial locus 1), in subjects with intermittent claudication (IC) secondary to peripheral arterial disease (PAD). VLTS-589 is an investigational nonviral therapeutic comprising a plasmid-expressing Del-1 formulated with poloxamer 188 (facilitating agent). One hundred subjects with bilateral PAD and IC will be randomized after careful screening to bilateral intramuscular delivery of VLTS-589 or placebo. A total of 84 mg of plasmid or placebo will be delivered as 42 intramuscular injections (2 ml per injection, 21 injections or 42 ml in each extremity of either plasmid or placebo) in both lower extremities. The subjects in the study will be followed at regular intervals for a year after study drug administration (days 30, 90, 180, and 365) with the primary endpoint being the safety and tolerability of VLTS-589 and change in peak walking time (PWT) at day 90. The secondary endpoints include percent and absolute change in resting ankle brachial Index, claudication onset time, and quality of life measured at various time points. DELTA-1 represents the largest plasmid-based gene transfer trial designed to test the efficacy of a Del-1 as a therapeutic approach in patients with IC caused by PAD. The novel aspects of the protocol include the usage of a Del-1 plasmid-polaxamer formulation to enhance gene transfer at doses that are an order of magnitude different than other comparable trials in a unique bilateral intramuscular dosing pattern to maximize transfection/clinical efficacy and general applicability to patients with PAD.
Collapse
Affiliation(s)
- Sanjay Rajagopalan
- Section of Vascular Medicine, Zena and Michael A. Wiener Cardiovascular Institute, Mount Sinai School of Medicine, Box #1030, One Gustave Levy Place, New York, NY 10029, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Barbash IM, Leor J, Feinberg MS, Tessone A, Aboulafia-Etzion S, Orenstein A, Ruiz-Cabello J, Cohen JS, Mardor Y. Interventional magnetic resonance imaging for guiding gene and cell transfer in the heart. BRITISH HEART JOURNAL 2004; 90:87-91. [PMID: 14676253 PMCID: PMC1768020 DOI: 10.1136/heart.90.1.87] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
BACKGROUND Interventional magnetic resonance imaging (iMRI) has the potential for guiding interventional cardiac procedures in real time. OBJECTIVES To test the feasibility of iMRI guided gene and cell transfer to the heart and to monitor myocardial remodelling after myocardial infarction in a rat model. METHODS The MRI contrast agent GdDTPA, together with either Evans blue dye, or a recombinant adenovirus encoding the LacZ gene, or primary fibroblasts tagged by BrdU, were injected into the myocardium of rats under iMRI guidance. Rats were killed seven days after the injection and the hearts sectioned to identify the blue dye, LacZ expression, or fibroblast presence, respectively. In a parallel study, left ventricular area was measured before and after myocardial infarction and in sham operated rats by T1 weighted MRI and by echocardiography. RESULTS Location of GdDTPA enhancement observed with iMRI at the time of injection was correlated with Evans blue stain, beta-gal expression, and the primary fibroblast location in histological studies. iMRI and echocardiography measured a comparable increase in left ventricular area at seven and 30 days after myocardial infarction. A good correlation was found between the iMRI and echocardiographic assessment of left ventricular area (r = 0.70; p < 0.0001) and change in left ventricular area with time (r = 0.75; p < 0.0001). CONCLUSIONS The results show the feasibility and efficiency of iMRI guided intramyocardial injections, and the ability to monitor heart remodelling using iMRI. Genes, proteins, or cells for tissue engineering could be injected accurately into the myocardial scar under iMRI guidance.
Collapse
Affiliation(s)
- I M Barbash
- Neufeld Cardiac Research Institute, Sheba Medical Centre, Tel-Hashomer, Israel
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Tanner FC, Largiadèr T, Greutert H, Yang Z, Lüscher TF. Nitric oxide synthase gene transfer inhibits biological features of bypass graft disease in the human saphenous vein. J Thorac Cardiovasc Surg 2004; 127:20-6. [PMID: 14752408 DOI: 10.1016/j.jtcvs.2003.07.021] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Bypass graft disease is related to proliferation and migration of vascular smooth muscle cells and to platelet activation with thrombus formation. Nitric oxide inhibits these biological responses; it has never been demonstrated, however, whether this occurs in intact human vascular tissue after endothelial nitric oxide synthase gene transfer. METHODS We examined whether endothelial nitric oxide synthase overexpression inhibits biological features of bypass graft disease in saphenous vein tissue. RESULTS The nitric oxide donor diethylenetriamineNONOate inhibited proliferation (P <.001) and migration (P <.001) of human saphenous vein vascular smooth muscle cells in response to 20% serum in a concentration-dependent manner. A similar effect on proliferation (P <.05) and migration (P <.05) without any cytotoxicity was observed after adenoviral endothelial nitric oxide synthase transfection. Staining of saphenous vein tissue for placental alkaline phosphatase demonstrated that adenoviral transfection was efficient. Consistent with this observation, endothelial nitric oxide synthase protein expression and nitric oxide release were enhanced in transfected tissue. Further, endothelial nitric oxide synthase overexpression inhibited vascular smooth muscle cell outgrowth from saphenous vein explants over 21 days; 48% +/- 12% of explants exhibited outgrowth after treatment with endothelial nitric oxide synthase adenovirus as compared with 69% +/- 10% in those infected with control adenovirus and 90% +/- 5% in uninfected tissue (P <.05). Similarly, platelet adhesion to human saphenous vein tissue was inhibited by endothelial nitric oxide synthase overexpression; adhesion was reduced in segments infected with endothelial nitric oxide synthase adenovirus (58% +/- 6%) as compared with those infected with control adenovirus (107% +/- 8%) or uninfected saphenous vein (100%; P <.05). CONCLUSIONS These data demonstrate that endothelial nitric oxide synthase gene transfer inhibits biological features of bypass graft disease in intact human saphenous vein tissue. Therefore, endothelial nitric oxide synthase transfection represents a promising gene transfer approach to prevent venous bypass graft disease.
Collapse
Affiliation(s)
- Felix C Tanner
- Cardiovascular Research, Clinic for Cardiovascular Surgery, Department of Clinical Research, University Hospital, Bern, Switzerland
| | | | | | | | | |
Collapse
|
32
|
Abstract
Gene therapy is an exciting frontier in medicine today. Many genes have been shown to be useful for treatment of various vascular diseases, including chronic cardiac and limb ischemia syndromes, vasculoproliferative disorder, hypercholesterolemia, atherosclerosis, thrombosis, and hypertension. Precise delivery of genes into target vessels, efficient transfer of genes into vascular cells of the target, and prompt assessment of gene expression over time are three challenging tasks for successful vascular gene therapy. Thus, in vivo imaging methods that can be used to monitor gene delivery and localize gene expression are needed. Modern imaging techniques provide an opportunity to monitor and direct vascular gene therapy. Radiologists play a key role not only in developing and mastering endovascular genetic interventions but also in assessing the success of vascular gene therapy and directing further refinement of vascular gene therapy technology. This article provides an overview of the current status of imaging of vascular gene therapy.
Collapse
Affiliation(s)
- Xiaoming Yang
- Department of Radiology, Johns Hopkins University School of Medicine, Traylor Bldg, Rm 330, 720 Rutland Ave, Baltimore, MD 21205, USA.
| |
Collapse
|
33
|
Avril N, Bengel FM. Defining the success of cardiac gene therapy: how can nuclear imaging contribute? Eur J Nucl Med Mol Imaging 2003; 30:757-71. [PMID: 12541135 DOI: 10.1007/s00259-002-1100-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Gene therapy is a promising modality for the treatment of various cardiovascular diseases such as ischaemia, heart failure, restenosis after revascularisation, hypertension and hyperlipidaemia. An increasing number of approaches are moving from experimental and preclinical validation to clinical application, and several multi-centre trials are currently underway. Despite the rapid progress in cardiac gene therapy, many basic tools and principles remain under development. Questions with regard to the optimal method for gene delivery in a given situation remain open, as do questions concerning therapeutic efficacy and the time course and magnitude of gene expression in target and remote areas. Nuclear imaging provides valuable tools to address these open issues non-invasively. Functional effects of molecular therapy at the tissue level can be identified using tracers of blood flow, metabolism, innervation or cell death. The use of reporter genes and radiolabelled reporter probes allows for non-invasive assessment of location, magnitude and persistence of transgene expression in the heart and the whole body. Co-expression of a reporter gene will allow for indirect imaging of the expression of a therapeutic gene of choice, and linkage of measures of transgene expression to downstream functional effects will enhance the understanding of basic mechanisms of cardiac gene therapy. Hence, nuclear imaging offers great potential to facilitate and refine the determination of therapeutic effects in preclinical and clinical cardiovascular gene therapy.
Collapse
Affiliation(s)
- Norbert Avril
- Division of Nuclear Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | | |
Collapse
|
34
|
Abstract
Vascular smooth muscle cell (VSMC) proliferation after arterial injury results in neointima formation and plays an important role in the pathogenesis of restenosis after angioplasty, in-stent restenosis, vascular bypass graft occlusion, and allograft vasculopathy. Major progress has been made recently in elucidating the cellular and molecular mechanisms underlying neointima formation. However, no known curative treatment currently exists. In cases in which pharmacologic and surgical interventions have had limited success, gene therapy remains a potential strategy for the treatment of such vascular proliferative diseases. To date, recombinant adenoviral vectors continue to be the most efficient methods of gene transfer into the arterial wall. However, concerns over the safety of using viral vectors in a clinical situation have inspired the considerable progress that has been made in improving both viral and nonviral modes of gene transfer. This review discusses some of the recent insights and outstanding progress in vascular gene therapeutic approaches to inhibit neointima both from a biologic and therapeutic perspective.
Collapse
Affiliation(s)
- Martin F Crook
- Cardiovascular Biology Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | | |
Collapse
|
35
|
Wentzel JJ, Aguiar SH, Fayad ZA. Vascular MRI in the diagnosis and therapy of the high risk atherosclerotic plaque. J Interv Cardiol 2003; 16:129-42. [PMID: 12768916 DOI: 10.1046/j.1540-8183.2003.08024.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Disruption of a high risk plaque is known as the primary cause of cardiovascular events. Characterization of arterial wall components has become an essential adjunct in the identification of patients with plaques prone to rupture. Magnetic Resonance Imaging (MRI) has been revealed as one of the noninvasive tools possibly capable of identifying and characterizing high risk atherosclerotic plaque. MRI may facilitate diagnosis, and guide and serially monitor interventional and pharmacological treatment of atherosclerotic disease. In addition, it permits the simultaneous assessment of the anatomy, morphology, and hemodynamics for the study of flow-induced atherogenesis. It possibly will identify asymptomatic patients with subclinical atherosclerosis. This has potential significance for the improvement of strategies in primary and secondary prevention.
Collapse
Affiliation(s)
- Jolanda J Wentzel
- Zena and Michael A. Wiener Cardiovascular Institute, Imaging Science Laboratories, Mount Sinai School of Medicine, New York, NY, USA
| | | | | |
Collapse
|
36
|
Chiu-Pinheiro CK, O'Brien T, Katusic ZS, Bonilla LF, Hamner CE, Schaff HV. Gene transfer to coronary artery bypass conduits. Ann Thorac Surg 2002; 74:1161-6; discussion 1166. [PMID: 12400762 DOI: 10.1016/s0003-4975(02)03831-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND Gene therapy is a rational approach to prevention of stenosis in saphenous vein grafts used as conduits for coronary artery bypass grafting. To explore this possibility we developed methods for adenoviral-mediated gene transfer to canine saphenous veins. METHODS During a single procedure, autogenous canine saphenous vein segments were transduced ex vivo and used as coronary artery bypass grafts. The proximal end of each vein was ligated, adenovirus containing the Escherichia coli beta-galactosidase gene (Ad.CMVLacZ) was delivered at titers of 2.5 x 10(9) or 5 x 10(9) plaque-forming units (pfu)/mL to the lumen through a distal heparin lock, and the segment was immersed in the viral solution for 1 hour at 37 degrees C. Control segments were exposed to diluent alone in an identical manner. Aortocoronary anastomoses were made using cardiopulmonary bypass. Transgene expression was assessed qualitatively and quantitatively after 3 days. RESULTS Beta-galactosidase levels showed a dose-dependent increase: 0.00 +/- 0.00 ng/mg total protein for controls; 5.60 +/- 2.27 ng/mg total protein for a viral titer of 2.5 x 10(9) pfu/mL and 11.97 +/- 6.14 ng/mg for 5 x 10(9) pfu/mL. The two dosage groups differed significantly from each other (p = 0.035) and from controls (p = 0.003). X-gal staining demonstrated mostly endothelial and scattered adventitial transgene expression. CONCLUSIONS Transgene expression after ex vivo gene transfer into saphenous vein grafts in a canine coronary artery bypass model indicates that this method may be useful for delivery of therapeutic genes to prevent or retard vein graft arteriosclerosis.
Collapse
|
37
|
Aikawa R, Huggins GS, Snyder RO. Cardiomyocyte-specific gene expression following recombinant adeno-associated viral vector transduction. J Biol Chem 2002; 277:18979-85. [PMID: 11889137 DOI: 10.1074/jbc.m201257200] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Recombinant adeno-associated viral (rAAV) vectors hold promise for delivering genes for heart diseases, but cardiac-specific expression by the use of rAAV has not been demonstrated. To achieve this goal rAAV vectors were generated expressing marker or potentially therapeutic genes under the control of the cardiac muscle-specific alpha myosin heavy chain (MHC) gene promoter. The rAAV-MHC vectors expressed in primary cardiomyocytes with similar kinetics to rAAV-CMV; however, expression by the rAAV-MHC vectors was restricted to cardiomyocytes. rAAV vectors have low cytotoxicity, and it is demonstrated here that rAAV fails to induce apoptosis in cardiomyocytes compared with a recombinant adenoviral vector. rAAV-MHC or rAAV-CMV vectors were administered to mice to determine the specificity of expression in vivo. The rAAV-MHC vectors expressed specifically in cardiomyocytes, whereas the control rAAV-CMV vector expressed in heart, skeletal muscle, and brain. rAAV-MHC transduction resulted in long term (16 weeks) expression of human growth hormone following intracardiac, yet not intramuscular, injection. Finally, we defined the minimal MHC enhancer/promoter sequences required for specific and robust in vivo expression in the context of a rAAV vector. For the first time we describe a panel of rAAV vectors capable of long term cardiac specific expression of intracellular and secreted proteins.
Collapse
Affiliation(s)
- Ryuichi Aikawa
- Cardiovascular Biology Laboratory, Harvard School of Public Health, Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | |
Collapse
|
38
|
Vannan M, McCreery T, Li P, Han Z, Unger E, Kuersten B, Nabel E, Rajagopalan S. Ultrasound-mediated transfection of canine myocardium by intravenous administration of cationic microbubble-linked plasmid DNA. J Am Soc Echocardiogr 2002; 15:214-8. [PMID: 11875383 DOI: 10.1067/mje.2002.119913] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We tested the hypothesis that targeted disruption of cationic microbubble-linked plasmid DNA, using diagnostic ultrasound, may aid transfection of large animal myocardium. Plasmid DNA encoding for CAT (pCAT, chloramphenicol acetyltransferase) was bound to a novel cationic microbubble containing MRX-225 for intravenous administration, and 16 dogs in 4 groups variously received this conjugate or plasmid only, or were exposed to ultrasound. Histochemical staining and enzyme-linked immunosorbent assay analysis showed CAT activity in the myocardium of only those animals that received microbubble-linked DNA and were exposed to ultrasound. Thus, disruption of cationic-linked, low-dose plasmid systems by diagnostic ultrasound may facilitate transfection of large animal hearts.
Collapse
Affiliation(s)
- Mani Vannan
- Department of Internal Medicine, Division of Cardiology, University of Michigan Health System, Ann Arbor, Michigan, USA.
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Abstract
Endothelium is an important target for drug or gene therapy because of its important role in the biological system. In this paper, we have characterized nanoparticle uptake by endothelial cells in cell culture. Nanoparticles were formulated using poly DL-lactide-co-glycolide polymer containing bovine serum albumin as a model protein and 6-coumarin as a fluorescent marker. It was observed that the cellular uptake of nanoparticles depends on the time of incubation and the concentration of nanoparticles in the medium. The uptake of nanoparticles was rapid with confocal microscopy demonstrating their localization mostly in the cytoplasm. The mitogenic study demonstrated biocompatability of nanoparticles with the cells. The study thus demonstrates that nanoparticles could be used for localizing therapeutic agents or gene into endothelial cells. Nanoparticles localized in the endothelium could provide prolonged drug effects because of their sustained release characterics, and also could protect the encapsulated agent from enzymatic degradation.
Collapse
Affiliation(s)
- Jasmine Davda
- Department of Pharmaceutical Sciences, College of Pharmacy, 986025 Nebraska Medical Center, Omaha, NE 68198-6025, USA
| | | |
Collapse
|
40
|
Vale PR, Isner JM, Rosenfield K. Therapeutic angiogenesis in critical limb and myocardial ischemia. J Interv Cardiol 2001; 14:511-28. [PMID: 12053643 DOI: 10.1111/j.1540-8183.2001.tb00367.x] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Research in animal models of ischemia has shown that administration of angiogenic growth factors, either as a recombinant protein or by gene transfer, can augment nutrient perfusion through neovascularization to promote the development of supplemental collateral blood vessels that will constitute endogenous bypass conduits around occluded native arteries; a strategy termed "therapeutic angiogenesis." In animal models and clinical trials, the best studied cytokines with angiogenic activity are vascular endothelial growth factor (VEGF) and fibroblast growth factor (FGF). Clinical trials of therapeutic angiogenesis in patients with critical limb ischemia demonstrated resolution of rest pain and/or improved limb integrity, increased pain-free walking time and ankle-brachial index, newly visible collateral vessels by digital subtraction angiography, and qualitative evidence of improved distal flow by magnetic resonance imaging. Initial clinical trials in patients with end-stage coronary artery disease using direct myocardial injection via thoracotomy resulted in large increases in exercise time and marked reductions in anginal symptoms, as well as objective evidence of improved perfusion and left ventricular function. Larger scale placebo-controlled trials have been limited to intracoronary and intravenous administration of recombinant protein, and have not shown significant improvement in exercise time or angina compared to placebo. Larger scale placebo-controlled studies of gene transfer using catheter-based endocardial delivery are in progress. Future clinical studies are required to determine the optimal dose, formulation, route of administration, and combinations of growth factors, as well as the requirement for endothelial progenitor cell or stem cell supplementation, to provide effective and safe therapeutic angiogenesis for patients with critical limb ischemia and chronic myocardial ischemia who are not candidates for conventional revascularization procedures.
Collapse
Affiliation(s)
- P R Vale
- Department of Vascular Medicine, St. Elizabeth's Medical Center, Tufts University School of Medicine, Boston, Massachusetts, USA
| | | | | |
Collapse
|
41
|
Vasquez EC, Meyrelles SS, Chapleau MW, Johnson AK. Approaches for gene delivery to the subfornical organ and magnocellular neurons. Clin Exp Pharmacol Physiol 2001; 28:602-9. [PMID: 11458888 DOI: 10.1046/j.1440-1681.2001.03492.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
- E C Vasquez
- Department of Physiological Sciences, Biomedical Center (UFES) and Health Sciences College (Emescam), Vitoria, Brazil
| | | | | | | |
Collapse
|
42
|
Abstract
This review surveys a range of approaches using plasmid DNA encoding the 165-amino-acid isoform of vascular endothelial growth factor (phVEGF165) to therapeutically modulate micro- or macrovascular endothelial cells, focusing on strategies to augment postnatal collateral circulation in arterial insufficiency or to accelerate re-endothelialization after balloon angioplasty to prevent restenosis. We focus on intra-arterial and intramuscular/intramyocardial gene transfer of the VEGF165 gene, the options that have been most thoroughly studied to date in patients. We review developmental and postnatal significance of the endothelial-cell-specific mitogen VEGF that has stimulated these studies and present limitations of current knowledge as well as challenges for the future.
Collapse
Affiliation(s)
- I Baumgartner
- Swiss Cardiovascular Center, Division Angiology, University Hospital, Bern, 3010 Switzerland.
| | | |
Collapse
|
43
|
Yang X, Liu H, Li D, Zhou X, Jung WC, Deans AE, Cui Y, Cheng L. Digital optical imaging of green fluorescent proteins for tracking vascular gene expression: feasibility study in rabbit and human cell models. Radiology 2001; 219:171-5. [PMID: 11274553 DOI: 10.1148/radiology.219.1.r01ap23171] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
PURPOSE To investigate the feasibility of using a sensitive digital optical imaging technique to detect green fluorescent protein (GFP) expressed in rabbit vasculature and human arterial smooth muscle cells. MATERIALS AND METHODS A GFP plasmid was transfected into human arterial smooth muscle cells to obtain a GFP-smooth muscle cell solution. This solution was imaged in cell phantoms by using a prototype digital optical imaging system. For in vivo validation, a GFP-lentivirus vector was transfected during surgery into the carotid arteries of two rabbits, and GFP-targeted vessels were harvested for digital optical imaging ex vivo. RESULTS Optical imaging of cell phantoms resulted in a spatial resolution of 25 microm/pixel. Fluorescent signals were detected as diffusely distributed bright spots. At ex vivo optical imaging of arterial tissues, the average fluorescent signal was significantly higher (P <.05) in GFP-targeted tissues (mean +/- SD, 9,357.3 absolute units of density +/- 1,001.3) than in control tissues (5,633.7 absolute units of density +/- 985.2). Both fluorescence microscopic and immunohistochemical findings confirmed these differences between GFP-targeted and control vessels. CONCLUSION The digital optical imaging system was sensitive to GFPs and may potentially provide an in vivo imaging tool to monitor and track vascular gene transfer and expression in experimental investigations.
Collapse
Affiliation(s)
- X Yang
- Department of Radiology, the Johns Hopkins University School of Medicine, Outpatient Center, Rm 4243, 601 N Caroline St, Baltimore, MD 21287-0845, USA.
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Abstract
The future of non-viral gene therapy depends on a detailed understanding of the barriers to delivery of polynucleotides. These include physicomechanical barriers, which limit the design of delivery devices, physicochemical barriers that influence self-assembly of colloidal particulate formulations, and biological barriers that compromise delivery of the DNA to its target site. It is important that realistic delivery strategies are adopted for early clinical trials in non-viral gene therapy. In the longer term, it should be possible to improve the efficiency of gene delivery by learning from the attributes which viruses have evolved; attributes that enable translocation of viral components across biological membranes. Assembly of stable, organized virus-like particles will require a higher level of control than current practice. Here, we summarize present knowledge of the biodistribution and cellular interactions of gene delivery systems and consider how improvements in gene delivery will be accomplished in the future.
Collapse
Affiliation(s)
- C W Pouton
- Department of Pharmacy and Pharmacology, University of Bath, Bath BA2 7AY, UK.
| | | |
Collapse
|
45
|
Abstract
Cardiomyocytes are terminally differentiated and are unable to proliferate in response to injury. Genetic modulation, cell transplantation and tissue engineering promise a revolutionary approach for myocardial regeneration and tissue repair after myocardial injury. Current data derived from animal models suggest that it may be possible to treat heart failure by inserting genetic materials or myogenic cells into injured myocardium. Success with animal models has raised the hope for new treatment after heart attacks and could prove an alternative to transplantation, particularly in elderly patients for whom there is often a lack of donor hearts. This exciting research, however, still faces significant difficulties before it can develop into a clinical therapeutic tool and many challenges need to be overcome before cell transplantation, gene therapy and tissue engineering can be considered efficient, therapeutic strategies for myocardial regeneration.
Collapse
Affiliation(s)
- S Etzion
- Neufeld Cardiac Research Institute, Tel-Aviv University, Sheba Medical Center, Tel-Hashomer, Israel
| | | | | | | |
Collapse
|
46
|
Wang Y, Boros P, Liu J, Qin L, Bai Y, Bielinska AU, Kukowska-Latallo JF, Baker JR, Bromberg JS. DNA/dendrimer complexes mediate gene transfer into murine cardiac transplants ex vivo. Mol Ther 2000; 2:602-8. [PMID: 11124061 DOI: 10.1006/mthe.2000.0201] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Starburst polyamidoamine dendrimers are synthetic polymers with unique structural and physical characteristics suitable for DNA gene transfer. Our previous studies demonstrated that Starburst dendrimers augment plasmid-mediated gene transfer efficiency in a nonvascularized, cardiac transplantation model. In this study, the fifth generation of ethylenediamine core dendrimer was investigated for its ability to enhance gene transfer and expression in a clinically relevant murine vascularized heart transplantation model. The plasmid pMP6A-beta-gal, encoding beta-galactosidase (beta-Gal), was incubated with dendrimers to form complexes. The complexes were perfused via the coronary arteries during donor graft harvesting, and reporter gene expression was determined by quantitative evaluation of X-Gal staining. The grafts infused with pMP6A-beta-gal/dendrimer complexes showed beta-Gal expression in myocytes from 7 to 14 days. A number of variables for transfer of the DNA/dendrimer complexes were tested, including DNA:dendrimer charge ratios, concentrations of DNA and dendrimer, preservation solutions, ischemic time, and enhancement of vascular permeability by serotonin, papaverine, and VEGF administration. The results showed that DNA/dendrimer complexes containing 20 microg of DNA and 260 microg of dendrimer (1:20 charge ratio) in a total volume of 200 microl resulted in highest gene expression in the grafts. The results also showed that prolonged incubation (cold ischemic time) to 2 h and pretreatment with serotonin further enhanced gene expression.
Collapse
Affiliation(s)
- Y Wang
- Institute for Gene Therapy and Molecular Medicine, Mount Sinai School of Medicine, New York, New York 10029, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Maillard L, Van Belle E, Tio FO, Rivard A, Kearney M, Branellec D, Steg PG, Isner JM, Walsh K. Effect of percutaneous adenovirus-mediated Gax gene delivery to the arterial wall in double-injured atheromatous stented rabbit iliac arteries. Gene Ther 2000; 7:1353-61. [PMID: 10981661 DOI: 10.1038/sj.gt.3301255] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Though the efficacy of intravascular gene transfer has been demonstrated in native vessels following acute injury, this methodology has not been validated in complex models of vascular injury that more closely mimic clinical angioplasty procedures. Previous studies have shown that Gax gene overexpression modulates the injury-induced remodeling of the vessel in rat carotid and normal rabbit iliac arteries. Here, we evaluated the effect of the Gax gene delivery in atheromatous stented vessels. Rabbits were fed 120 g daily of 1% cholesterol diet for 3 weeks. At 1 week they underwent initial injury on the external iliac artery, then balloon angioplasty was performed at 3 weeks at the same site with a 2.5 mm diameter channel balloon catheter (three times 1 min at 6 atm). Either saline (n = 4) or the control viral construct Ad-CMVluc (5 x 109 p.f.u.) (n = 5) or Ad-CMVGax (5 x 10(9) p.f.u.) (n = 4) was delivered with a poloxamer mixture via a channel balloon (6 atm, 30 min), and a 15 mm long Palmaz-Schatz stent (PS154) was then deployed at the site (1 min, 8 atm). Arteries were analyzed 1 month later. At 1 month, the Ad-CMVGax treated arteries exhibited a lower maximal intimal area (1. 15+/-0.1 mm2) than saline (1.87+/-0.15 mm2, P = 0.007) or Ad-CMVluc-treated vessels (1.98+/-0.31 mm2, P = 0.04). Likewise Ad-CMVGax-treated vessels displayed a lower maximal percentage cross-sectional area narrowing (35.1+/-3.5%) than saline (65.3+/-9.4%, P = 0.01) or Ad-CMVluc-treated vessels (62.7+/-6.7%, P = 0.02). Angiographic analysis revealed larger minimal lumen diameter in Ad-CMVGax treated arteries (2.0+/-0.1 mm) than saline (1.14+/-0.36 mm, P = 0.06) or Ad-CMVluc-treated vessels (1.23+/-0.25 mm, P = 0.02). Overexpression of the Gax gene inhibits neointimal hyperplasia and lumen loss in atheromatous stented rabbit iliac arteries.
Collapse
Affiliation(s)
- L Maillard
- Cardiologie A, D et USCI, Laboratoire de Biotechnologie et Génétique Expérimentale, Hôpital Trousseau, Tours, France
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Toyoda K, Andresen JJ, Zabner J, Faraci FM, Heistad DD. Calcium phosphate precipitates augment adenovirus-mediated gene transfer to blood vessels in vitro and in vivo. Gene Ther 2000; 7:1284-91. [PMID: 10918499 DOI: 10.1038/sj.gt.3301214] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Adenovirus (Ad)-mediated gene transfer to blood vessels is relatively inefficient, probably because binding of adenovirus to the endothelium and adventitia seems to be limited. Association of calcium phosphate (CaPi) precipitates with adenovirus improves efficiency of gene transfer to some cells in culture and to mouse lung in vivo. In this study, we tested the hypothesis that CaPi is useful for adenovirus-mediated gene transfer to blood vessels. In fibroblast and endothelial cells in culture, Ad:CaPi coprecipitates greatly increased transgene expression. Ad:CaPi also enhanced transgene expression in both adventitia and endothelium of carotid arteries and aortae from rabbits studied ex vivo. After injection of Ad:CaPi into the cisterna magna of rabbits in vivo, the transgene product was markedly increased in leptomeninges of the ventral brain stem, including the adventitia of the basilar artery. We also examined mechanisms of enhanced gene transfer. Binding of adenovirus to fibroblast and endothelial cells in culture, and to the basilar artery in vivo, as determined using Southern blot analysis, was augmented by CaPi. Antibody to adenoviral fiber knob did not inhibit augmented transgene expression by Ad:CaPi. The finding suggests that improved adenoviral binding occurs primarily via a fiber-independent pathway. Thus, CaPi precipitates are useful for improvement of adenovirus-mediated gene transfer to blood vessels in vitro and in vivo.
Collapse
Affiliation(s)
- K Toyoda
- Department of Internal Medicine, Pharmacology, University of Iowa College of Medicine, and Veterans Administration Medical Center, Iowa City 52242, USA
| | | | | | | | | |
Collapse
|
49
|
Abrahams JM, Diamond SL, Hurst RW, Zager EL, Grady MS. Topic review: surface modifications enhancing biological activity of guglielmi detachable coils in treating intracranial aneurysms. SURGICAL NEUROLOGY 2000; 54:34-40; discussion 40-1. [PMID: 11024505 DOI: 10.1016/s0090-3019(00)00269-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND Endovascular therapy with Guglielmi detachable coils is an accepted treatment option for patients with intracranial aneurysms. However, an emerging technology in the realm of endovascular tools is the use of traditional Guglielmi detachable coils with biologically active substances complexed to the coil surface to enhance aneurysm occlusion. METHODS We review the literature and current trends in modified Guglielmi detachable coils. Surface modifications with extracellular matrix proteins, growth factors, ion impregnation, and genetically altered cells have been used in animal studies to improve the cellular response of Guglielmi detachable coils. Similarly, coronary artery stents have been modified in several different ways to maintain vessel patency, contrary to the goal of endovascular therapy. We comparatively reviewed this literature to add insight into the evolution of the research on modified Guglielmi detachable coils. CONCLUSIONS Guglielmi detachable coil modifications have the potential to enhance aneurysm obliteration with directed cellular responses. This may allow aneurysm occlusion with coils in less time than untreated coils, thus decreasing the risks of aneurysm enlargement and hemorrhage.
Collapse
Affiliation(s)
- J M Abrahams
- Department of Neurosurgery, The Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | | | | | | | | |
Collapse
|
50
|
Marshall DJ, Palasis M, Lepore JJ, Leiden JM. Biocompatibility of cardiovascular gene delivery catheters with adenovirus vectors: an important determinant of the efficiency of cardiovascular gene transfer. Mol Ther 2000; 1:423-9. [PMID: 10933963 DOI: 10.1006/mthe.2000.0059] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Gene therapy approaches hold promise for the treatment of a wide variety of cardiovascular diseases. Many strategies for cardiovascular gene therapy involve catheter-mediated vector delivery via intramyocardial injection, intracoronary infusion, or direct gene transfer into the vessel wall. Several different gene delivery catheters have been developed and utilized in preclinical and clinical studies of cardiovascular gene therapy. However, rigorous studies of the biocompatibility of these catheters with gene therapy vectors have not yet been reported. In this report, we have examined the compatibility of cardiovascular gene therapy catheters and catheter constituents with first-generation E1/E3-deleted adenovirus vectors. We show that (i) currently available catheters rapidly and efficiently inactivate adenovirus vector infectivity; (ii) this inactivation is mediated by a variety of commonly used catheter constituents including stainless steel, nitinol, and polycarbonate; (iii) catheter-mediated inactivation of adenovirus vectors can be prevented by preflushing catheters with solutions of serum albumin; and (iv) it is possible to identify a set of catheter materials that are compatible with current adenovirus vectors. These results underscore the importance of catheter/vector compatibility and suggest methods for increasing the efficiency of catheter-mediated cardiovascular gene therapy.
Collapse
Affiliation(s)
- D J Marshall
- Harvard School of Public Health, Boston, Massachusetts 02115, USA
| | | | | | | |
Collapse
|