1
|
Zhao Q, Liu F, Zhao Q, Zhang J, Luo J, Li X, Yang Y. Constitutive activation of ERK1/2 signaling protects against myocardial ischemia via inhibition of mitochondrial fragmentation in the aging heart. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:479. [PMID: 33850876 PMCID: PMC8039677 DOI: 10.21037/atm-21-503] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Background Studies have shown that the ability of the myocardium to tolerate ischemia becomes significantly compromised with age. During ischemia, several endogenous protective signals are activated to protect the heart from injury, among which extracellular-signal regulated kinase (ERK) 1/2 signaling has been established as playing a pivotal role. However, in aging hearts, the activation of ERK1/2 is compromised. Mitogen-activated protein kinase/ERK kinase (MEK) is a major regulator of ERK1/2 signaling. In the present study, we investigated whether transduction of CaMEK, a constitutively activated MEK, using adeno-associated virus serotype 9 (AAV9) could protect the aging heart against ischemia. Methods Myocardial ischemia models were established in aging mice and senescent cardiomyocytes, and AAV9-mediated delivery of CaMEK was applied. Echocardiography, fluorescent staining, transmission electron microscopy, flow cytometry, and immunoblotting were used to explore the effects of CaMEK and their underlying mechanism. Results AAV9-CaMEK activated ERK1/2 signaling and exerted cardioprotective effects against ischemia in aging hearts. Specifically, CaMEK transduction decreased dynamin-related protein-1 (Drp1) expression and phosphorylation at serine 616, resulting in improved mitochondrial morphology and function in aging ischemic hearts. Furthermore, CaMEK transduction exerted similar protective effects in senescent cardiomyocytes under hypoxia. Meanwhile, with the inhibition of ERK1/2 signaling in senescent cardiomyocytes under hypoxia, the opposite effects were observed, including an increase in mitochondrial fragmentation and aggravation of mitochondrial dysfunction and cell apoptosis. Conclusions Our results suggested that AAV9-CaMEK alleviated ischemia-induced myocardium injury in the aging heart, at least in part, through inhibition of mitochondrial fragmentation. Therefore, AAV9-CaMEK is a potential intervention for prevention of ischemia-induced injury of the aging myocardium.
Collapse
Affiliation(s)
- Qiang Zhao
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Fen Liu
- Xinjiang Key Laboratory of Cardiovascular Disease Research, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Qian Zhao
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Jinyu Zhang
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Junyi Luo
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Xiaomei Li
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Yining Yang
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China.,Department of Cardiology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| |
Collapse
|
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
|
Hangalapura BN, Timares L, Oosterhoff D, Scheper RJ, Curiel DT, de Gruijl TD. CD40-targeted adenoviral cancer vaccines: the long and winding road to the clinic. J Gene Med 2012; 14:416-27. [PMID: 22228547 DOI: 10.1002/jgm.1648] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
The ability of dendritic cells (DCs) to orchestrate innate and adaptive immune responses has been exploited to develop potent anti-cancer immunotherapies. Recent clinical trials exploring the efficacy of ex vivo modified autologous DC-based vaccines have reported some promising results. However, in vitro generation of autologous DCs for clinical administration, their loading with tumor associated antigens (TAAs) and their activation, is laborious and expensive, and, as a result of inter-individual variability in the personalized vaccines, remains poorly standardized. An attractive alternative approach is to load resident DCs in vivo by targeted delivery of TAAs, using viral vectors and activating them simultaneously. To this end, we have constructed genetically-modified adenoviral (Ad) vectors and bispecific adaptor molecules to retarget Ad vectors encoding TAAs to the CD40 receptor on DCs. Pre-clinical human and murine studies conducted so far have clearly demonstrated the suitability of a 'two-component' (i.e. Ad and adaptor molecule) configuration for targeted modification of DCs in vivo for cancer immunotherapy. This review summarizes recent progress in the development of CD40-targeted Ad-based cancer vaccines and highlights pre-clinical issues in the clinical translation of this approach.
Collapse
Affiliation(s)
- Basav N Hangalapura
- Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam, The Netherlands
| | | | | | | | | | | |
Collapse
|
4
|
Feng M, He ZM, Zhu YX, Liu LH, Lu CW, Xiong Y. Improvement of endothelial dysfunction in atherosclerotic rabbit aortas by ex vivo gene transferring of dimethylarginine dimethylaminohydrolase-2. Int J Cardiol 2010; 144:180-6. [DOI: 10.1016/j.ijcard.2010.04.019] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2009] [Revised: 01/25/2010] [Accepted: 04/03/2010] [Indexed: 10/19/2022]
|
5
|
Abstract
The introduction of the heart lung machine more than 50 years ago proved in principle that heart function can be replaced, albeit for short periods. This was followed by attempts to produce total or partial artificial hearts that could function for prolonged periods of time. Progress in this field has been intermittent but has accelerated considerably in the past 10 years, with ventricular assist devices (VADs) reaching an impressive degree of sophistication and complexity owing to the contributions from clinicians, engineers, scientists, industrialists, and others. This review describes the currently available types of VADs, their current clinical use, the patient selection process, the trend toward use of VADs in patients with less severe heart failure, and the use of VADs for myocardial recovery in combination with novel pharmacological strategies, gene therapy, and cell therapy.
Collapse
Affiliation(s)
- Cesare M Terracciano
- Harefield Heart Science Centre, Imperial College London, Harefield, Middlesex UB9 6JH, UK
| | | | | |
Collapse
|
6
|
Shen C, Gu M, Song C, Miao L, Hu L, Liang D, Zheng C. The tumorigenicity diversification in human embryonic kidney 293 cell line cultured in vitro. Biologicals 2008; 36:263-8. [PMID: 18378163 DOI: 10.1016/j.biologicals.2008.02.002] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2007] [Revised: 02/12/2008] [Accepted: 02/21/2008] [Indexed: 11/25/2022] Open
Abstract
The Human Embryonic Kidney (HEK) 293 cell line is widely used in research work such as vaccine production, adenovirus and adeno-associated viral vectors, and gene therapy. However, little attention was drawn to the passage level of 293 cells. We first claim that the tumorigenicity of the HEK 293 cell line reached 100% when the passage exceeded 65, whereas using low-passage (<52) HEK 293 cell line no tumor could be induced under the same condition. Results from nude mice assay, tumor tissue histological examination, primary culture, PCR and isoenzyme analysis showed that the tumor in nude mice could only be induced by viable high-passage 293 cells. This suggests that more attention should be paid to the passage level of the HEK 293 cell line, especially for vaccine production but the low-passage HEK 293 cell line should be acceptable to regulatory authorities for recombinant virus vector, vaccines, and gene therapy. Meanwhile, we also find that high-passage HEK 293 can be employed as a highly malignant tumor model as its tumorigenicity increases significantly.
Collapse
Affiliation(s)
- Chao Shen
- State key laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | | | | | | | | | | | | |
Collapse
|
7
|
Abstract
Molecular cardiology is a new and fast-growing area of cardiovascular medicine that aims to apply molecular biology techniques for the mechanistic investigation, diagnosis, prevention and treatment of cardiovascular disease. As an emerging discipline, it has changed conceptual thinking of cardiovascular development, disease etiology and pathophysiology. Although molecular cardiology is still at a very early stage, it has opened a promising avenue for understanding and controlling cardiovascular disease. With the rapid development and application of molecular biology techniques, scientists and clinicians are closer to curing heart diseases that were thought to be incurable 20 years ago. There clearly is a need for a more thorough understanding of the molecular mechanisms of cardiovascular diseases to promote the advancement of stem cell therapy and gene therapy for heart diseases. The present paper briefly reviews the state-of-the-art techniques in the following areas of molecular cardiology: gene analysis in the diseased heart; transgenic techniques in cardiac research; gene transfer and gene therapy for cardiovascular disease; and stem cell therapy for cardiovascular disease.
Collapse
Affiliation(s)
- Zhongjie Sun
- Department of Medicine, College of Medicine, University of Florida, Gainesville 32610-0274, USA.
| |
Collapse
|
8
|
Wang X, Sun Z, Cade R. Prolonged attenuation of cold-induced hypertension by adenoviral delivery of renin antisense. Kidney Int 2005; 68:680-7. [PMID: 16014045 DOI: 10.1111/j.1523-1755.2005.00446.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND Renin has been linked to the pathogenesis of some forms of hypertension, including cold-induced hypertension (CIH). Although several antihypertensive drugs that inhibit angiotensin-converting enzyme (ACE) and angiotensin II (Ang II) type 1 (AT(1)) receptors are available, they are short-lasting and have side effects. Inhibition of renin [the first and rate-limiting step of the renin-angiotensin system (RAS)] would provide an inhibition of the entire RAS. Thus, we developed an antisense approach for specific inhibition of renin based on the genetic design. The objective of this study was to test our hypothesis that adenoviral delivery of renin antisense inhibits renin and attenuates CIH. METHODS Recombinant adenoviruses carrying rat renin antisense (rAdv.RRA) and LacZ reporter gene (rAdv.LacZ) were constructed and used for in vivo gene transfer via intravenous injection. Four groups of rats were used (six rats/group). Blood pressure did not differ among the four groups during the control period at room temperature (25 degrees C). Two groups of rats received rAdv.RRA (2.5 x 10(9) pfu/rat, intravenously), while the other two groups received the same dose of rAdv.LacZ and served as controls. After gene delivery, one rAdv.LacZ-treated and one rAdv.RRA-treated group were exposed to cold (5 degrees C), while the remaining groups were kept at 25 degrees C. Blood pressure was monitored weekly during cold exposure. A 24-hour urine sample was collected during weeks 1, 3, and 5 for measuring urinary aldosterone excretion. At the end of week 5, all animals were killed and blood was collected for measurement of plasma renin activity (PRA), total plasma renin, plasma active renin, and plasma aldosterone. Vascular Ang II contents were measured in all rats. RESULTS Blood pressure of the rAdv.LacZ-treated group rose significantly within 2 weeks of exposure to cold and reached 158.2 +/- 6.4 mm Hg by week 5. In contrast, blood pressure (117.1 +/- 5.3 mm Hg) of the cold-exposed group treated with rAdv.RRA did not increase until 5 weeks after exposure to cold. Thus, a single dose of rAdv.RRA prevented CIH for at least 5 weeks. rAdv.RRA abolished the cold-induced increases in PRA, total plasma renin, plasma active renin, vascular Ang II, and plasma and urine aldosterone, indicating effective inhibition of the entire RAS. CONCLUSION rAdv.RRA effectively inhibited the entire RAS and produced prolonged attenuation of CIH. Antisense inhibition of renin may be a novel and ideal approach for long-term control of hypertension.
Collapse
Affiliation(s)
- Xiuqing Wang
- Department of Medicine, College of Medicine, University of Florida, Gainesville, Florida 32610, USA
| | | | | |
Collapse
|
9
|
Ramsden JD, Buchanan MA, Egginton S, Watkinson JC, Mautner V, Eggo MC. Complete inhibition of goiter in mice requires combined gene therapy modification of angiopoietin, vascular endothelial growth factor, and fibroblast growth factor signaling. Endocrinology 2005; 146:2895-902. [PMID: 15817662 DOI: 10.1210/en.2005-0168] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
In goiter, increased expression of growth factors and their receptors occurs. We have inhibited the action of some of these growth factors, alone and in combination, to determine which are important in goitrogenesis. Recombinant adenovirus vectors (RAds) expressing truncated, secreted forms of human Tie2 (RAd-sTie2) and vascular endothelial growth factor receptor 1 (RAd-sVEGFR1) or a truncated, dominant-negative fibroblast growth factor receptor 1 (RAdDN-FGFR1) were used. Goiters in mice were induced by feeding an iodide-deficient diet, containing methimazole and sodium perchlorate. RAds were administered to mice simultaneously with the goitrogenic regimen, which was continued for 14 d. RAd treatment did not significantly affect increases in TSH or reductions in thyroid hormone or thyroid hyperactivity seen in goitrogen-treated controls mice, suggesting no effect on pituitary or thyroid responses to hypothyroidism. In control goiters, a 4-fold increase in vascular volume accompanied a 2-fold increase in thyroid mass. Complete inhibition of these increases was found when animals were treated with the three RAds in combination. In thyroids from three RAd-treated animals, there was marked, significant inhibition of Tie2, FGFR1, VEGFR1, FGF-2, and VEGF expression, compared with control goiters. When used individually, RAdDN-FGFR1 partially prevented goiter and RAd-sVEGFR1 partially reduced vascular volume. Their effects were not additive. RAd-sTie2 did not reduce goiter mass or vascular volume when used alone but was essential for complete goiter inhibition. VEGF and VEGFR1 expression was reduced in these thyroids. Limitation of physiologic organ growth is complex, requiring inhibition of multiple, interdependent growth factor axes.
Collapse
Affiliation(s)
- James D Ramsden
- Division of Medical Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | | | | | | | | | | |
Collapse
|
10
|
|
11
|
Wang X, Cade R, Sun Z. Expression of human eNOS in cardiac and endothelial cells. METHODS IN MOLECULAR MEDICINE 2005; 112:91-107. [PMID: 16010013 DOI: 10.1007/978-1-59259-879-3_7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
In this chapter we provide a detailed technique-focused protocol for expression of human endothelial nitric oxide synthase (eNOS) gene delivered by replication-deficient adenovirus. It includes construction of recombinant plasmid with human eNOS gene full-length cDNA (heNOS), packaging of adenovirus with heNOS (Adv-heNOS) in 293 cells, confirmation of Adv-heNOS, amplification of high-titer stockAdv-heNOS, determination of the Adv-heNOS titer, expression of Adv-heNOS in the human abdominal aorta endothelial (HAAE1) cell line and rat heart myoblast cell line (H9C2), and measurement of human eNOS activity and NO production.
Collapse
Affiliation(s)
- Xiuqing Wang
- Department of Medicine, University of Florida, Gainesville, FL, USA
| | | | | |
Collapse
|
12
|
Akowuah EF, Sheridan PJ, Cooper GJ, Newman C. Preventing saphenous vein graft failure: does gene therapy have a role? Ann Thorac Surg 2003; 76:959-66. [PMID: 12963247 DOI: 10.1016/s0003-4975(03)00505-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Gene therapy potentially allows local delivery and expression of cytokines, growth factors, and other mediators. In spite of increasing knowledge of the human genome, applications in clinical practice are only just beginning. The main limitations of effective clinical gene therapy are safety and low transfection efficiency. Saphenous vein grafts permit the transfection of the conduit ex vivo. This allows a variety of transfection techniques to be used, enhancing the transfection efficiency while limiting the risk of systemic complications. This review examines the potential mechanisms of gene delivery and genetic targets that may be applied to saphenous vein graft failure.
Collapse
Affiliation(s)
- Enoch F Akowuah
- Cardiovascular Research Group, The University of Sheffield, Sheffield, United Kingdom.
| | | | | | | |
Collapse
|
13
|
Ooboshi H, Ibayashi S, Takada J, Kumai Y, Iida M. Brain ischemia as a potential target of gene therapy. Exp Gerontol 2003; 38:183-7. [PMID: 12543276 DOI: 10.1016/s0531-5565(02)00150-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Brain infarction is one of the most important age-associated medical conditions, and the age-related neuronal vulnerability to brain ischemia is suggested to play an important role. Recent advancements in gene transfer techniques have provided promising approaches to the treatment of brain ischemia. In experimental studies, the ischemic penumbra area can be targeted by gene transfer even after ischemic insult, and post-ischemic gene therapy seems effective in attenuation of ischemic damage in both global and focal brain ischemia. Perivascular approaches of gene transfer to the cerebral blood vessels through the subarachnoid space may lead to prevention of brain ischemia caused by vasospasm after subarachnoid hemorrhage. Gene transfer to cerebral blood vessels and ischemic brain tissue may offer future therapeutic approaches to stroke.
Collapse
Affiliation(s)
- Hiroaki Ooboshi
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan.
| | | | | | | | | |
Collapse
|
14
|
Shin HK, Lee JH, Kim CD, Kim YK, Hong JY, Hong KW. Prevention of impairment of cerebral blood flow autoregulation during acute stage of subarachnoid hemorrhage by gene transfer of Cu/Zn SOD-1 to cerebral vessels. J Cereb Blood Flow Metab 2003; 23:111-20. [PMID: 12500096 DOI: 10.1097/01.wcb.0000036561.60552.63] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The present study determined whether gene transfer of human copper/zinc superoxide dismutase-1 (Cu/Zn SOD-1) prevented the autoregulatory impairment of CBF induced by subarachnoid hemorrhage (SAH). After application of recombinant adenovirus (100 microL of 1 x 10(10) pfu/mL, intracisternally) encoding human Cu/Zn SOD-1 3 days before experiments, Cu/Zn SOD-1 activity significantly increased in association with increase in Cu/Zn SOD-1 mRNA and protein expression in the cerebral vasculature of both sham-operated and SAH rats as determined by reverse transcriptase-polymerase chain reaction, Western blotting, and immunohistochemistry, and SAH-induced increase in superoxide anion was markedly reduced in accordance with increased nitric oxide production. In line with these findings, rats that received human Cu/Zn SOD-1 therapy showed the prominent restoration of blunted vasodilation of the pial artery in response to calcitonin gene-related peptide and levcromakalim, and the recovery of impaired autoregulatory vasodilation in response to acute hypotension, thereby leading to significant restoration of CBF autoregulation. These results provide a rational basis for application of Cu/Zn SOD-1 gene therapy for protection of the impairment of autoregulatory CBF during the acute stage of SAH.
Collapse
Affiliation(s)
- Hwa Kyoung Shin
- Department of Pharmacology and Internal Medicine, College of Medicine, Research Institute of Genetic Engineering, Pusan National University, Seo-Gu, Busan, South Korea
| | | | | | | | | | | |
Collapse
|
15
|
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
|
16
|
Kim CD, Shin HK, Lee HS, Lee JH, Lee TH, Hong KW. Gene transfer of Cu/Zn SOD to cerebral vessels prevents FPI-induced CBF autoregulatory dysfunction. Am J Physiol Heart Circ Physiol 2002; 282:H1836-42. [PMID: 11959650 DOI: 10.1152/ajpheart.00590.2001] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The goal of this study was to determine whether gene transfer of human copper-zinc (Cu/Zn) superoxide dismutase (SOD) has preventive effects on cerebral blood flow (CBF) autoregulatory dysfunction after fluid percussion injury (FPI). Rats subjected to FPI (2-2.5 atm) exhibited enhanced activity of reduced NADP (NADPH) oxidase in the cerebral vasculature. In line with these findings, the rats showed not only reduced vasodilation of the pial artery in response to calcitonin gene-related peptide and levcromakalim but also impaired autoregulatory vasodilation in response to acute hypotension. The FPI-induced hemodynamic alterations were significantly prevented by pretreatment with diphenyleneiodonium (10 micromol/l), an NAD(P)H oxidase inhibitor. Intracisternal application of recombinant adenovirus (100 microl of 1 x 10(10) pfu/ml)-encoding human Cu/Zn SOD 3 days before FPI prevented the impairment of vasodilation to hypotension and vasorelaxants, resulting in the restoration of CBF autoregulation. Our findings demonstrate that FPI-induced impairment of CBF autoregulation is closely related with NAD(P)H oxidase-derived superoxide anion, and these alterations can be prevented by the recombinant adenovirus-mediated transfer of human Cu/Zn SOD gene to the cerebral vasculature.
Collapse
Affiliation(s)
- Chi Dae Kim
- Department of Pharmacology, College of Medicine, Pusan National University, Pusan 602-739, South Korea
| | | | | | | | | | | |
Collapse
|
17
|
|
18
|
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
|
19
|
Hingtgen SD, Davisson RL. Gene therapeutic approaches to oxidative stress-induced cardiac disease: principles, progress, and prospects. Antioxid Redox Signal 2001; 3:433-49. [PMID: 11491655 DOI: 10.1089/15230860152409077] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Heart and vascular diseases continue to rank among the most frequent and devastating disorders to affect adults in many parts of the world. Increasing evidence from a variety of experimental models indicates that reactive oxygen species can play a key role in the development of myocardial damage from ischemia/reperfusion, the development of cardiac hypertrophy, and the transition of hypertrophy to cardiac failure. The recent dramatic increase in availability of genomic data has included information on the genetic modulation of reactive oxygen species and the antioxidant systems that normally prevent damage from these radicals. Nearly simultaneously, progressively more sophisticated and powerful methods for altering the genetic complement of selected tissues and cells have permitted application of gene therapeutic methods to understand better the pathophysiology of reactive oxygen species-mediated myocardial damage and to attenuate or treat that damage. Although exciting and promising, gene therapy approaches to these common disorders are still in the experimental and developmental stages. Improved understanding of pathophysiology, better gene delivery systems, and specific gene therapeutic strategies will be needed before gene therapy of oxyradical-mediated myocardial damage becomes a clinical reality.
Collapse
Affiliation(s)
- S D Hingtgen
- Department of Anatomy and Cell Biology, College of Medicine, The University of Iowa, Iowa City 52242, USA
| | | |
Collapse
|
20
|
Yap J, Pellegrini C, O'Brien T, Tazelaar HD, McGregor CG. Conditions of vector delivery improve efficiency of adenoviral-mediated gene transfer to the transplanted heart. Eur J Cardiothorac Surg 2001; 19:702-7. [PMID: 11343956 DOI: 10.1016/s1010-7940(01)00673-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
OBJECTIVES Conditions for ex vivo gene transfer to the transplanted heart were studied in a model of syngeneic abdominal heterotopic heart transplantation in the rat. Various methods of adenoviral-mediated gene transfer to the transplanted heart were compared. METHODS In the first experiment, a dose response study, an adenoviral vector encoding the beta-galactosidase gene was infused into the donor heart with the pulmonary artery open and flushed out prior to performing the transplant. In the second experiment, the effects of clamping the pulmonary artery during vector infusion and not flushing out the viral solution, resulting in vector dwell during the warm ischemia, were examined. RESULTS In the first experiment, gene transfer was relatively inefficient; however, transgene expression improved with increases in the vector dose (range, 1x10(7)-1x10(9)). The efficiency of gene transfer was significantly greater when the conditions of the second experiment were applied. In all models studied, cardiomyocytes and not vascular endothelial cells were the predominant cell type transduced. CONCLUSIONS This study indicates that the conditions of adenoviral vector delivery are critical for optimizing gene transfer in the transplant setting. In addition, intravascular administration of adenoviral vector to the donor heart results predominantly in cardiomyocyte transgene expression.
Collapse
Affiliation(s)
- J Yap
- Department of Surgery, Mayo Clinic and Foundation, Rochester, MN 55905, USA
| | | | | | | | | |
Collapse
|
21
|
Medina-Kauwe LK, Kasahara N, Kedes L. 3PO, a novel nonviral gene delivery system using engineered Ad5 penton proteins. Gene Ther 2001; 8:795-803. [PMID: 11420644 DOI: 10.1038/sj.gt.3301448] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2000] [Accepted: 01/31/2001] [Indexed: 11/09/2022]
Abstract
This study describes the development of 3PO, a nonviral, protein-based gene delivery vector which utilizes the highly evolved cell-binding, cell-entry and intracellular transport functions of the adenovirus serotype 5 (Ad5) capsid penton protein. A penton fusion protein containing a polylysine sequence was produced by recombinant methods and tested for gene delivery capability. As the protein itself is known to bind integrins through a conserved consensus motif, the penton inherently possesses the ability to bind and enter cells through receptor-mediated internalization. The ability to lyse the cellular endosome encapsulating internalized receptors is also attributed to the penton. The recombinant protein gains the additional function of DNA binding and transport with the appendage of a polylysine motif. This protein retains the ability to form pentamers and mediates delivery of a reporter gene to cultured cells. Interference by oligopeptides bearing the integrin binding motif suggests that delivery is mediated specifically through integrin receptor binding and internalization. The addition of protamine to penton-DNA complexes allows gene delivery in the presence of serum.
Collapse
Affiliation(s)
- L K Medina-Kauwe
- Institute for Genetic Medicine and Department of Biochemistry and Molecular Biology, University of Southern California Keck School of Medicine, Los Angeles, CA 90033, USA
| | | | | |
Collapse
|
22
|
Abstract
Cardiovascular[TRACE;del] disease is the leading cause of death in the US and world-wide. Advances in molecular biology and the human genome project have revealed opportunities for novel strategies for cardiac gene therapy. This review discusses general and specific aspects of gene transfer strategies in cardiac tissues. These include 1) the selection and/or optimization of the vector for gene transfer; 2) the identification of the target gene(s); 3) the use of cardiac-specific promoters; and 4) the use of an appropriate delivery system for administration. Currently, several vectors (e.g., viral and nonviral vectors) have been developed and many target genes have been identified (e.g., VEGF, FGF, beta-AR, etc.). Many investigations have provided experimental models for gene delivery systems but the most efficient cardiac gene transfer was obtained from intramyocardial injection or perfusion of explanted myocardium. The data available thus far have suggested favorable immediate effects following gene transfer, but long-term value of cardiac gene therapy has not been proven. Further refinements in appropriate vectors that provide cell or tissue selectivity and long-lasting effects are necessary as well as the development of minimally invasive procedures for gene transfer.
Collapse
Affiliation(s)
- S K Wattanapitayakul
- Department of Pharmacology, Faculty of Medicine, Srinakharinwirot University, Bangkok, Thailand
| | | |
Collapse
|
23
|
Shinmura K, Morishita R, Aoki M, Higaki J, Ogihara T, Kaneda Y, Tani M. Catheter-delivered in vivo gene transfer into rat myocardium using the fusigenic liposomal mediated method. JAPANESE HEART JOURNAL 2000; 41:633-47. [PMID: 11132170 DOI: 10.1536/jhj.41.633] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
We compared the efficacy of four different in vivo hemagglutinating virus of Japan (HVJ)-liposome gene transfer methods, i.e., direct myocardial injection (i.m.), injection into the left ventricular cavity (LV), infusion at the level of the coronary cusps (CI), or injection into the left ventricular cavity with a balloon catheter blocking aortic flow (LV+B) to transfer beta-galactosidase, FlTC-labeled oligodeoxynucleotide (ODN), and/or luciferase genes into the rat heart. I.m. caused highly efficient gene transfer in the limited area around the injection site, which suggests that i.m. may be a suitable method for targeted treatment of focal lesion. In the LV+B group, all rats had myocardial beta-galactosidase staining and fluorescence of FITC-labeled ODN in the nuclei of cardiac myocytes around the coronary arteries and the vasa vasorum, and some transfected myocytes were observed in the middle of the myocardium without any evidence of injury. In contrast, in the CI group, only half of the animals had myocardial expression of beta-galactosidase. In contrast, fluorescence or luciferase activity was present throughout the left ventricle in the LV+B group. However, the percentage of myocytes that exhibited fluorescence was less than 1% of the total ventricular myocyte population and luciferase activity in the LV+B group was 1.6% of that in the i.m. group. No evidence of luciferase expression was observed in brain, lung, liver, kidney, or testis in either the i.m. or LV+B group. These results suggest that HVJ-liposome gene transfer into the myocardium through the coronary arteries using a balloon-catheter technique is safe and has the potential for causing widespread transgene expression with organ-specificity, although the efficiency of gene transfer should be improved.
Collapse
Affiliation(s)
- K Shinmura
- Department of Geriatric Medicine, Keio University School of Medicine, Tokyo, Japan
| | | | | | | | | | | | | |
Collapse
|
24
|
O'Brien T, Simari RD. Gene therapy for atherosclerotic cardiovascular disease: a time for optimism and caution. Mayo Clin Proc 2000; 75:831-4. [PMID: 10943238 DOI: 10.4065/75.8.831] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Cardiovascular disease is the leading cause of death in the Western world, and gene therapy approaches to several cardiovascular disorders have been proposed. One of the major stumbling blocks to be overcome before widespread clinical use of this technology is how to deliver DNA efficiently and safely to cells in vivo. While delivery of DNA alone is inefficient, use of viral vectors may overcome this problem. Adenoviral vectors are most commonly used in cardiovascular gene delivery, but toxicity related to these vectors remains a concern. In addition, duration of gene expression with use of these vectors is limited, which may be advantageous in settings in which transient expression is satisfactory to obtain a therapeutic effect. Gene therapy has been suggested as an approach to multiple conditions, including restenosis after angioplasty, therapeutic neovascularization, and bypass graft restenosis. Phase 1 clinical trials were recently reported. While proof of principle has been established in preclinical animal models, convincing efficacy data in humans do not yet exist. Improvements in vector technology and methods of catheter-mediated vascular gene delivery are needed before widespread clinical application of this therapy.
Collapse
Affiliation(s)
- T O'Brien
- Division of Endocrinology, Metabolism, and Nutrition and Internal Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | | |
Collapse
|
25
|
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
|
26
|
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
|
27
|
Palasis M, Luo Z, Barry JJ, Walsh K. Analysis of adenoviral transport mechanisms in the vessel wall and optimization of gene transfer using local delivery catheters. Hum Gene Ther 2000; 11:237-46. [PMID: 10680838 DOI: 10.1089/10430340050015987] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Local delivery devices have been used for adenovirus-mediated gene transfer to the arterial wall for the potential treatment of vascular proliferative diseases. However, low levels of adenoviral gene expression in vascular smooth muscle cells may pose a serious limitation to the success of these procedures in the clinic. In this study, we examined the mechanisms controlling adenoviral transport to the vessel wall, using both hydrogel-coated and infusion-based local delivery catheters, with the goal of enhancing in vivo gene transfer under clinically relevant delivery conditions. The following delivery parameters were tested in vivo: applied transmural pressure, viral solution volume and concentration, and delivery time. We found that viral particles are transported into the vessel wall in a manner consistent with diffusion rather than pressure-driven convection. Consistent with diffusion, viral concentration was shown to be the key variable for viral transport in the vessel wall and thus gene expression in vascular smooth muscle cells. A transduction level of 17.8+/-3.2% was achieved by delivering a low volume of concentrated adenoviral beta-galactosidase solution through an infusion balloon catheter at low pressure without an adverse effect on medial cellularity. Under these conditions, effective gene transfer was accomplished within a clinically relevant time frame of 2 min, indicating that longer delivery times may not be necessary to achieve efficient gene transfer.
Collapse
Affiliation(s)
- M Palasis
- Boston Scientific Corporation, Natick, MA 01760, USA.
| | | | | | | |
Collapse
|
28
|
Abstract
Complexing recombinant DNA with cationic liposomes is a convenient means of introducing foreign genes into cells (lipofection) and could potentially form the basis for genetically modifying diseased blood vessels in patients. The mechanism of lipofection is incompletely understood, but it is recognized that the degree of successful gene transfer is highly dependent on cell type. We have transfected primary cultures of human vascular smooth muscle cells with a plasmid expressing either firefly luciferase (Luc) or nuclear-localized beta-galactosidase (NL-beta-gal). Cells were derived from either normal human internal mammary arteries, fragments of primary atherosclerotic plaque, or fragments of restenotic lesion. Concurrent lipofection of rabbit vascular smooth muscle cells and NIH 3T3 cells was performed as well. Compared with NIH 3T3 cells, expression in human vascular smooth muscle cells was markedly reduced: In cells derived from internal mammary artery, Luc expression, normalized for protein content, was 123-fold lower than in NIH 3T3 cells, while the proportion of cells expressing NL-beta-gal was 30-fold lower. Luc expression in cells derived from restenotic tissue was significantly greater than from cells derived from primary plaque. Within a given population of cells, the mitotic index of cells expressing the recombinant gene was significantly higher than the mitotic index for the total population of cells (p < 0.05). Finally, cotransfection experiments, in which lipofection of smooth muscle cells was performed using genes for NL-beta-gal and for human growth hormone, showed that among positive transfectants a high proportion of cells (23-36%) coexpressed both genes. Thus, the efficiency of successful lipofection in human vascular smooth muscle cells in vitro is low. Transfection appears to be preferentially facilitated in cells derived from restenotic tissue and specific properties of smooth muscle cells, including growth rates, appear to be critical for successful transfection. Further elucidation of cell properties that promote transfection is required to augment the efficiency of liposome-mediated gene transfer in human vascular cells.
Collapse
|
29
|
Christenson SD, Lund D, Ooboshi H, Faraci FM, Davidson L, Heistad DD. Approaches to enhance expression after adenovirus-mediated gene transfer to the carotid artery. ENDOTHELIUM : JOURNAL OF ENDOTHELIAL CELL RESEARCH 1999; 7:75-82. [PMID: 10599561 DOI: 10.3109/10623329909165313] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The goal of this study was to enhance transgene expression after adenoviral-mediated gene transfer to the carotid artery. We used an adenoviral vector with a transgene that expresses beta-galactosidase, driven by the human cytomegalovirus (CMV) promoter/enhancer. The CMV promoter drives constitutive expression, and response elements within the enhancer allow inducible expression through binding of active transcription factors, such as cAMP response element binding protein (CREB) and nuclear factor kappa B (NFkappaB). Rings of rabbit carotid artery were incubated ex vivo with a replication-deficient adenovirus that expresses beta-galactosidase (AdCMV-betagal). Virus was removed from the medium, and forskolin or phorbol-12-myristate-13-acetate (PMA), which can induce activation of CREB or NFkappaB, respectively, were added to the medium. Pyrrolidine dithiocarbamate (PDTC) was used to inhibit activation of NFkappaB. Following incubation for 24 hours, beta-galactosidase activity was assessed by chemiluminescent reporter assay. Forskolin and PMA enhanced transgene expression in the carotid artery. Activity increased from 56+/-13 mU/mg protein (mean+/-SE) in rings of carotid treated with virus alone (10(9) pfu) to 159+/-23 mU/mg protein (P<0.05) in rings treated with forskolin, and to 189+/-40 mU/mg protein (P<0.05) in rings treated with PMA. Phorbol didecanoate, an inactive phorbol, did not affect expression of beta-galactosidase. After pre-incubation with PDTC prior to PMA, expression of beta-galactosidase was less than in rings incubated with PMA alone (29+/-11, P<0.05). Histochemical staining of carotid artery for beta-galactosidase demonstrated enhanced endothelial expression following administration of PMA. These findings suggest that expression after gene transfer to the carotid artery using an adenoviral vector with the CMV promoter/enhancer may be enhanced by PMA and forskolin, perhaps by activation of transcription factors.
Collapse
Affiliation(s)
- S D Christenson
- Department of Internal Medicine and Center on Aging University of Iowa College of Medicine and Veterans Administration Medical Center Iowa City, 52242, USA
| | | | | | | | | | | |
Collapse
|
30
|
Aschner JL, Kovacs N, Perciaccante JV, Figueroa JP, Thrikawala N, Robins GS, Busija DW. Endothelial nitric oxide synthase gene transfer enhances dilation of newborn piglet pulmonary arteries. THE AMERICAN JOURNAL OF PHYSIOLOGY 1999; 277:H371-9. [PMID: 10409217 DOI: 10.1152/ajpheart.1999.277.1.h371] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We determined the expression and functional correlate of in vitro transfection with a recombinant adenoviral vector encoding the gene for bovine endothelial nitric oxide synthase (AdCMVeNOS) or Escherichia coli beta-galactosidase (AdCMVLacZ) in pulmonary endothelial cells (EC), vascular smooth muscle cells (VSMC), and pulmonary arteries (PA) from newborn piglets. AdCMVeNOS and AdCMVeLacZ vectors, grown in 293-cell monolayers, were purified by double-cesium gradient ultracentrifugation. Cell cultures and PA were incubated with increasing vector titers for 30 or 60 min, followed by incubation in fresh medium for 18 h at 37 degrees C. LacZ expression was assessed by histochemical staining; eNOS expression was evaluated by Western blot analysis. Functional eNOS expression was determined by measurement of cGMP and quantification of the relaxation response to bradykinin (BK). In PA, LacZ transgene expression was preferentially localized to the adventitia and endothelium. Increased eNOS protein expression was observed in EC and VSMC transfected with AdCMVeNOS. Functional studies revealed increased cGMP abundance in cultured cells and enhanced relaxation to BK in AdCMVeNOS-transfected PA. These studies demonstrate that gene transfer with AdCMVeNOS results in functional expression and altered vasoactive responses in the neonatal pulmonary vasculature. Gene transfer with replication-deficient adenovirus vectors is a useful tool for the study of targeted genes in vascular biology.
Collapse
Affiliation(s)
- J L Aschner
- Department of Pediatrics, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157-1081, USA.
| | | | | | | | | | | | | |
Collapse
|
31
|
Abstract
Recent advances in the field of molecular biology have led to a better understanding of the pathological mechanisms of cardiovascular disease. The impact of these findings will shape the future of treatment modalities for cardiovascular disorders. Postulated targets and biological rationale of new techniques are being developed in a race towards molecular therapies for vascular diseases. Whether it is modulation of transmembrane cell receptors or phenotypic changes via vectors that mediate gene transfer, there is no doubt that molecular strategies will be an integral part of the future. Here we examine past and recent perspectives, describe directions and challenges in cardiac and cardiovascular areas of research, and discuss relevance to the field of cardiovascular perfusion.
Collapse
Affiliation(s)
- A Basile-Borgia
- Department of Cardiovascular Perfusion, Easton Hospital, Pennsylvania 18042, USA.
| | | | | |
Collapse
|
32
|
Rosenberg SA, Zhai Y, Yang JC, Schwartzentruber DJ, Hwu P, Marincola FM, Topalian SL, Restifo NP, Seipp CA, Einhorn JH, Roberts B, White DE. Immunizing patients with metastatic melanoma using recombinant adenoviruses encoding MART-1 or gp100 melanoma antigens. J Natl Cancer Inst 1998; 90:1894-900. [PMID: 9862627 PMCID: PMC2249697 DOI: 10.1093/jnci/90.24.1894] [Citation(s) in RCA: 206] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND The characterization of the genes encoding melanoma-associated antigens MART-1 or gp100, recognized by T cells, has opened new possibilities for the development of immunization strategies for patients with metastatic melanoma. With the use of recombinant adenoviruses expressing either MART-1 or gp100 to immunize patients with metastatic melanoma, we evaluated the safety, immunologic, and potential therapeutic aspects of these immunizations. METHODS In phase I studies, 54 patients received escalating doses (between 10(7) and 10(11) plaque-forming units) of recombinant adenovirus encoding either MART-1 or gp100 melanoma antigen administered either alone or followed by the administration of interleukin 2 (IL-2). The immunologic impact of these immunizations on the development of cellular and antibody reactivity was assayed. RESULTS Recombinant adenoviruses expressing MART-1 or gp100 were safely administered. One of 16 patients with metastatic melanoma receiving the recombinant adenovirus MART-1 alone experienced a complete response. Other patients achieved objective responses, but they had received IL-2 along with an adenovirus, and their responses could be attributed to the cytokine. Immunologic assays showed no consistent immunization to the MART-1 or gp100 transgenes expressed by the recombinant adenoviruses. High levels of neutralizing antibody were found in the pretreatment sera of the patients. CONCLUSIONS High doses of recombinant adenoviruses could be safely administered to cancer patients. High levels of neutralizing antibody present in patients' sera prior to treatment may have impaired the ability of these viruses to immunize patients against melanoma antigens.
Collapse
Affiliation(s)
- S A Rosenberg
- Surgery Branch, National Cancer Institute, Bethesda, MD, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Vasquez EC, Johnson RF, Beltz TG, Haskell RE, Davidson BL, Johnson AK. Replication-deficient adenovirus vector transfer of gfp reporter gene into supraoptic nucleus and subfornical organ neurons. Exp Neurol 1998; 154:353-65. [PMID: 9878173 DOI: 10.1006/exnr.1998.6917] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The present studies used defined cells of the subfornical organ (SFO) and supraoptic nuclei (SON) as model systems to demonstrate the efficacy of replication-deficient adenovirus (Ad) encoding green fluorescent protein (GFP) for gene transfer. The studies investigated the effects of both direct transfection of the SON and indirect transfection (i.e., via retrograde transport) of SFO neurons. The SON of rats were injected with Ad (2 x 10(6) pfu) and sacrificed 1-7 days later for cell culture of the SON and of the SFO. In the SON, GFP fluorescence was visualized in both neuronal and nonneuronal cells while only neurons in the SFO expressed GFP. Successful in vitro transfection of cultured cells from the SON and SFO was also achieved with Ad (2 x 10(6) to 2 x 10(8) pfu). The expression of GFP in in vitro transfected cells was higher in nonneuronal (approximately 28% in SON and SFO) than neuronal (approximately 4% in SON and 10% in SFO) cells. The expression of GFP was time and viral concentration related. No apparent alterations in cellular morphology of transfected cells were detected and electrophysiological characterization of transfected cells was similar between GFP-expressing and nonexpressing neurons. We conclude that (1) GFP is an effective marker for gene transfer in living SON and SFO cells, (2) Ad infects both neuronal and nonneuronal cells, (3) Ad is taken up by axonal projections from the SON and retrogradely transported to the SFO where it is expressed at detectable levels, and (4) Ad does not adversely affect neuronal viability. These results demonstrate the feasibility of using adenoviral vectors to deliver genes to the SFO-SON axis.
Collapse
Affiliation(s)
- E C Vasquez
- Department of Psychology, Cardiovascular Center, University of Iowa, Iowa City, Iowa, 52242, USA
| | | | | | | | | | | |
Collapse
|
34
|
Inesi G, Wade R, Rogers T. The sarcoplasmic reticulum Ca2+ pump: inhibition by thapsigargin and enhancement by adenovirus-mediated gene transfer. Ann N Y Acad Sci 1998; 853:195-206. [PMID: 10603947 DOI: 10.1111/j.1749-6632.1998.tb08267.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The role of the sarcoplasmic reticulum Ca2+ pump in the excitation-contraction coupling of cardiac muscle fibers was evaluated in experiments on SR ATPase inhibition with thapsigargin or, alternatively, on Ca2+ pump enhancement by SR ATPase transgenic expression. We found that thapsigargin, a highly specific and potent inhibitor of the SR ATPase, produces a strong reduction of cytosolic Ca2+ transient and contractile activation in neonatal rat myocytes, in the absence of any other functional effect. On the other hand, Ca2+ pump enhancement by ATPase transgenic expression affects dramatically Ca2+ transient and twitches, resulting in shorter duration and more rapid decay rates. Of particular interest is gene transfer mediated by recombinant adenovirus vectors under control of a cell-specific promoter, resulting in transgenic expression of all myocytes in culture, and no expression in fibroblasts.
Collapse
Affiliation(s)
- G Inesi
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore 21201, USA
| | | | | |
Collapse
|
35
|
Rios CD, Chu Y, Davidson BL, Heistad DD. Ten steps to gene therapy for cardiovascular diseases. THE JOURNAL OF LABORATORY AND CLINICAL MEDICINE 1998; 132:104-11. [PMID: 9708571 DOI: 10.1016/s0022-2143(98)90005-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Affiliation(s)
- C D Rios
- Department of Internal Medicine, the Cardiovascular Center, University of Iowa College of Medicine, Iowa City, USA
| | | | | | | |
Collapse
|
36
|
Chen AF, O'Brien T, Katusic ZS. Transfer and expression of recombinant nitric oxide synthase genes in the cardiovascular system. Trends Pharmacol Sci 1998; 19:276-86. [PMID: 9703761 DOI: 10.1016/s0165-6147(98)01190-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Gene therapy involves the transfer of a functional gene into host cells to correct the malfunction of a specific gene or to alleviate the symptoms of a disease. For gene transfer to the cardiovascular system, adenoviral vectors are the most efficient means of transfer. Recently, transfer and functional expression of recombinant nitrio oxide synthase (NOS) genes to cerebral and cardiovascular beds have been demonstrated both ex vivo and in vivo. Here, Alex Chen and colleagues review current progress in the field of vascular NOS gene transfer and the potential use of NOS gene therapy for a number of cardiovascular diseases. Although the feasibility of the NOS gene transfer approach has been demonstrated in animal models, currently available vectors have a number of technical and safety limitations that have to be solved before human NOS gene therapy for cardiovascular disease can be attempted.
Collapse
Affiliation(s)
- A F Chen
- Department of Anesthesiology, Mayo Clinic, Rochester, MN 55905, USA
| | | | | |
Collapse
|
37
|
Van Belle E, Maillard L, Rivard A, Fabre JE, Couffinhal T, Kearney M, Branellec D, Feldman LJ, Walsh K, Isner JM. Effects of poloxamer 407 on transfection time and percutaneous adenovirus-mediated gene transfer in native and stented vessels. Hum Gene Ther 1998; 9:1013-24. [PMID: 9607413 DOI: 10.1089/hum.1998.9.7-1013] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
UNLABELLED Reduction in transfection time and the ability to perform gene transfer in conjunction with endovascular stent implantation constitute two important challenges for percutaneous adenovirus-mediated gene transfer to vessel walls. Studies have suggested that the use of biocompatible polyol poloxamer 407 could be useful. We first evaluated the use of poloxamer 407 for percutaneous gene transfer in nonstented rabbit iliac arteries. A 200-microl mixture of Ad-RSVbetagal or Ad-CMVLuc in either phosphate-buffered saline (PBS) or 20% poloxamer was delivered. After 3 days, gene transfection was evaluated by X-Gal staining or measurement of luciferase activity. Poloxamer use resulted in a 3- to 15-fold increase in the percentage of transfected cells (X-Gal, p = 0.001) and a 16-fold increase in protein product (luciferase activity, p = 0.03), and allowed a decrease in transfection time from 30 to 5 min with minimal reduction in transfection efficiency. We then evaluated the feasibility of percutaneous gene transfer, using Ad-RSVbetagal diluted in pure PBS or 20% poloxamer, in conjunction with stent implantation. Gene delivery was performed either immediately before (pre-) or after (post-) stent implantation. When adenoviruses were diluted in PBS, gene transfer had a low efficiency (prestent, 0.3%; poststent, 0.2%; NS). With poloxamer, the efficacy was much higher (p = 0.0001) and similar "pre" (2.2%) or "post" (1.7%) stent delivery (NS). CONCLUSIONS (1) The use of poloxamer, rather than PBS, as a vehicle increases the efficacy of percutaneous adenovirus-mediated gene transfer and reduces transfection time; (2) gene transfer performed during stent implantation with poloxamer is feasible and achieves a significant level of gene expression. Thus percutaneous gene delivery is applicable to conventional stents and could present an attractive method by which to achieve local biological effects in a stent environment.
Collapse
Affiliation(s)
- E Van Belle
- Department of Medicine (Cardiology), St. Elizabeth's Medical Center, Tufts University School of Medicine, Boston, MA 02135, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Schneider DB, Fly CA, Dichek DA, Geary RL. Adenoviral gene transfer in arteries of hypercholesterolemic nonhuman primates. Hum Gene Ther 1998; 9:815-21. [PMID: 9581904 DOI: 10.1089/hum.1998.9.6-815] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Arterial gene transfer with adenoviral vectors is a promising approach for the treatment and prevention of vascular disorders. However, in small animals such as rats and rabbits adenoviral vectors can have deleterious effects on the artery wall. The effects of adenovirus in primate arteries have not been studied. AdRSVn-LacZ, a replication-defective adenoviral vector, was delivered to the left brachial arteries of six hypercholesterolemic cynomolgus monkeys; right brachial arteries received vehicle only. Serum was collected before gene transfer and at vessel harvest 9 or 10 days later. Recombinant gene expression was present in occasional endothelial cells of transduced arteries, and all animals generated neutralizing antibodies. In transduced arteries, immunostaining revealed a fourfold increase in intimal and medial macrophage accumulation (p < 0.05); intimal cellularity was also significantly increased (twofold; p < 0.05). T cell density and total cellular proliferation (determined by bromodeoxyuridine labeling) were unaffected. In hypercholesterolemic nonhuman primates, adenoviral vectors increase vessel wall inflammation and promote the progression of early atherosclerotic lesions. The long-term consequences of these observations remain unclear; however, a better understanding of host responses to specific vector systems appears necessary for the development of safe and effective approaches to human vascular gene therapy.
Collapse
Affiliation(s)
- D B Schneider
- Gladstone Institute of Cardiovascular Disease, Department of Surgery, University of California, San Francisco 94143, USA
| | | | | | | |
Collapse
|
39
|
Jeppsson A, Lee R, Pellegrini C, O'Brien T, Tazelaar HD, McGregor CG. Gene therapy in lung transplantation: effective gene transfer via the airways. J Thorac Cardiovasc Surg 1998; 115:638-43. [PMID: 9535452 DOI: 10.1016/s0022-5223(98)70329-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVES Gene therapy may provide a means of modifying factors that contribute to the development of pathologic processes in transplanted lungs. Experiments were designed to study the feasibility of adenovirus-mediated gene transfer by way of the airways to the transplanted lung. METHODS Orthotopic left lung transplantation (Lewis to Lewis rats) was performed on four groups of animals. 300 microl of adenovirus solution encoding for beta-galactosidase was infused into the left bronchus of donor rats at viral concentrations of 10(8) pfu/ml (n = 5), 10(9) pfu/ml (n = 6), and 10(10) pfu/ml (n = 6), and the lung was ventilated for 5 minutes. Controls (n = 6) received medium only. Seven days after transplantation, native and transduced, transplanted lungs were harvested. Sections of lung were fixed and stained with a solution of X-Gal (5-bromo-4-chloro-3-indolyl-beta-D-galactopyranoside) and staining was evaluated for distribution by cell type and intensity. RESULTS Beta-galactosidase expression was absent in the control group and in the native lungs. Two of five lungs in the 10(8) group expressed beta-galactosidase, but in a limited distribution and intensity. All six lungs in the 10(9) group and five of six lungs in the 10(10) group expressed beta-galactosidase. The distribution and intensity of beta-galactosidase expression ranged from only a few cells staining per slide to up to 75%. Pneumocytes were the most frequently stained cell type followed by alveolar macrophages. CONCLUSIONS Gene transfer to the transplanted lung via the bronchial route is feasible and offers a novel technique to modify pathologic processes in the transplanted lung.
Collapse
Affiliation(s)
- A Jeppsson
- Department of Surgery, Mayo Clinic and Foundation, Rochester, Minn 55905, USA
| | | | | | | | | | | |
Collapse
|
40
|
Inesi G, Lewis D, Sumbilla C, Nandi A, Strock C, Huff KW, Rogers TB, Johns DC, Kessler PD, Ordahl CP. Cell-specific promoter in adenovirus vector for transgenic expression of SERCA1 ATPase in cardiac myocytes. THE AMERICAN JOURNAL OF PHYSIOLOGY 1998; 274:C645-53. [PMID: 9530095 DOI: 10.1152/ajpcell.1998.274.3.c645] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Adenovirus-mediated transfer of cDNA encoding the chicken skeletal muscle sarco(endo)plasmic reticulum Ca(2+)-ATPase (SERCA1) yielded selective expression in cultured chick embryo cardiac myocytes under control of a segment (-268 base pair) of the cell-specific cardiac troponin T (cTnT) promoter or nonselective expression in myocytes and fibroblasts under control of a constitutive viral [cytomegalovirus (CMV)] promoter. Under optimal conditions nearly all cardiac myocytes in culture were shown to express transgenic SERCA1 ATPase. Expression was targeted to intracellular membranes and was recovered in subcellular fractions with a pattern identical to that of the endogenous SERCA2a ATPase. Relative to control myocytes, transgenic SERCA1 expression increased up to four times the rates of ATP-dependent (and thapsigargin-sensitive) Ca2+ transport activity of cell homogenates. Although the CMV promoter was more active than the cTnT promoter, an upper limit for transgenic expression of functional enzyme was reached under control of either promoter by adjustment of the adenovirus plaque-forming unit titer of infection media. Cytosolic Ca2+ concentration transients and tension development of whole myocytes were also influenced to a similar limit by transgenic expression of SERCA1 under control of either promoter. Our experiments demonstrate that a cell-specific protein promoter in recombinant adenovirus vectors yields highly efficient and selective transgene expression of a membrane-bound and functional enzyme in cardiac myocytes.
Collapse
Affiliation(s)
- G Inesi
- Department of Anatomy, University of California School of Medicine, San Francisco 94143, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Hogaboam CM, Vallance BA, Kumar A, Addison CL, Graham FL, Gauldie J, Collins SM. Therapeutic effects of interleukin-4 gene transfer in experimental inflammatory bowel disease. J Clin Invest 1997; 100:2766-76. [PMID: 9389741 PMCID: PMC508481 DOI: 10.1172/jci119823] [Citation(s) in RCA: 100] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Inflammatory bowel disease (IBD) is characterized by altered immunoregulation and augmented intestinal synthesis of nitric oxide. The purpose of this study was to determine the effects of exogenous IL-4, introduced by a recombinant human type 5 adenovirus (Ad5) vector, on the tissue injury associated with an experimental model of colonic immune activation and inflammation. Colitis was induced in rats by the intrarectal administration of trinitrobenzene sulfonic acid (TNB) dissolved in 50% ethanol, and control rats received saline via the same route. 1 h later, all rats were randomized into two groups. The first group was injected intraperitoneally (ip) with 3.0 x 10(6) plaque forming units (PFUs) of Ad5 transfected with murine interleukin-4 (Ad5IL-4) and the second group was injected ip with the same amount of Ad5 expressing the Escherichia coli Lac Z gene (Ad5LacZ). One-half of the colitic and control rats were injected again with 3.0 x 10(6) PFUs of Ad5IL-4 or Ad5LacZ on day 3 of the 6-d study. When introduced once or twice via the peritoneal route into control rats, Ad5LacZ was localized to the serosal lining of the peritoneal cavity, the diaphragm and the liver on day 6. One or two injections of Ad5IL-4 into rats also produced measurable levels of circulating IL-4. TNB-colitis in both Ad5LacZ-treated groups was associated with pronounced elevations in serum IFN-gamma, and mucosal ulceration of the distal colon. Myeloperoxidase and inducible nitric oxide synthase II (NOS II) synthetic activity were also increased by 30- and fivefold, respectively, above control levels in the distal colon. However, two injections of Ad5IL-4 into colitic rats caused the overexpression of IL-4, and significantly inhibited tissue damage, serum and colon IFN-gamma levels and myeloperoxidase activity in the distal colon. In addition, NOS II gene expression and NOS II nitric oxide synthesis was significantly inhibited. No therapeutic effect was observed in rats injected once with Ad5IL-4. Thus, IL-4, introduced by Ad5, is therapeutic during acute inflammation in the rat colon. The therapeutic effect of IL-4 was associated with an inhibition of inducible nitric oxide expression and a reduction in nitric oxide synthesis.
Collapse
Affiliation(s)
- C M Hogaboam
- Intestinal Disease Research Programme, McMaster University, Hamilton, Ontario, Canada.
| | | | | | | | | | | | | |
Collapse
|
42
|
Brauner R, Nonoyama M, Laks H, Drinkwater DC, McCaffery S, Drake T, Berk AJ, Sen L, Wu L. Intracoronary adenovirus-mediated transfer of immunosuppressive cytokine genes prolongs allograft survival. J Thorac Cardiovasc Surg 1997; 114:923-33. [PMID: 9434687 DOI: 10.1016/s0022-5223(97)70006-0] [Citation(s) in RCA: 50] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Intracoronary transfer and expression of recombinant genes in the intact heart is now feasible. In the transplant setting, local modulation of host immune responses by a genetically modified allograft may offer an attractive alternative to systemic immunosuppression. METHODS We tested the efficacy and in vivo effect of intracoronary transfer of two immunosuppressive cytokine genes. First-generation E1-deleted adenoviral vectors expressing the Epstein-Barr virus interleukin-10 (AdSvIL10) or human transforming growth factor--beta 1 (AdCMVTGF-beta) were used. Rabbit cardiac allografts were transduced during cold preservation by slow (1 ml/min) intracoronary infusion of 10(10) pfu/gm diluted viral vectors and then implanted heterotopically. Controls included E1-deleted adenovirus (Ad5dI434) and AdCMVLacZ. Beating allografts were collected on day 4 for analysis of gene transfer efficacy and distribution. Additional grafts were used for evaluation of alloreactivity (n = 34). RESULTS Mean allograft viral uptake was 81% (up to 91%). Polymerase chain reactions and reverse transcription-polymerase chain reactions confirmed the presence and expression of both genes in the grafts. beta-Galactosidase staining in AdCMVLacZ-infected grafts demonstrated efficient gene expression, which was highest (100%) in subepicardial regions. More homogeneous transmyocardial distribution of the transgene (in 25% to 40% of cells) could be achieved by pulsatile slow delivery. Allograft survival was 6.9 +/- 0.9 days in controls (n = 12), 11.1 +/- 1.7 days in AdCMVTGF-beta-infected grafts (n = 11, p < 10(-4)), and 11.2 +/- 3 days in AdSvIL10-infected grafts (n = 11, p < 10(-4)). Histologic scores (blinded) showed significantly (p < 0.005) higher regression coefficients for rejection in controls compared with both cytokine-transduced groups. Perioperative administration of cyclosporine A (INN: ciclosporin) to recipients had no effect on survival of AdCMVTGF-beta-infected grafts but reduced survival of AdSvIL10-infected grafts. CONCLUSIONS Intracoronary gene transfer of immunosuppressive cytokines to cardiac allografts is efficient and effectively prolongs graft survival. Vectors that would induce long-term expression of such genes may make this approach clinically applicable.
Collapse
Affiliation(s)
- R Brauner
- Division of Cardiothoracic Surgery, UCLA Medical Center 90095, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Meyrelles SS, Sharma RV, Whiteis CA, Davidson BL, Chapleau MW. Adenovirus-mediated gene transfer to cultured nodose sensory neurons. BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH 1997; 51:33-41. [PMID: 9427504 DOI: 10.1016/s0169-328x(97)00212-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Recent advances have enabled transfer of genes to various types of cells and tissues. The goals of the present study were to transfer genes to nodose sensory neurons using replication-deficient adenovirus vectors and to define the conditions needed to optimize the gene transfer. Neurons were dissociated from rat nodose ganglia and maintained in culture. Cultures were exposed for 30 min to vectors containing the beta-galactosidase gene lacZ driven by either the Rous sarcoma virus (RSV) or the cytomegalovirus (CMV) promoter. Cultures were fixed and treated with X-gal to evaluate lacZ expression 1-7 days after exposure to virus. Increasing concentrations of virus led to dose-related increases in the number of neurons expressing lacZ. LacZ was expressed in 8 +/- 2, 39 +/- 6, and 82 +/- 3% of neurons 1 day after exposure to 10(7), 10(8), and 10(9) pfu/ml of AdRSVlacZ, respectively (P < 0.05). The same doses of AdCMVlacZ led to expression in 41 +/- 9, 60 +/- 10, and 86 +/- 4% of neurons. Expression driven by the CMV promoter was essentially maximal within 1 day and remained stable for at least 7 days. In contrast, expression driven by the RSV promoter was less on day 1 but increased over time (1-7 days). There was no lacZ expression in vehicle-treated cultures and exposure to the adenovirus vectors did not adversely influence cell viability. Exposure of the neuronal cultures to an adenovirus vector containing the gene for green fluorescent protein (AdRSVgfp, 10(9) pfu/ml) enabled visualization of successful gene transfer in living neurons. The results indicate that gene transfer to cultured nodose neurons can be accomplished using adenovirus vectors. The expression of the transferred gene persists for at least 7 days, occurs more rapidly when expression is driven by the CMV compared with the RSV promoter, and occurs without adversely affecting cell viability.
Collapse
Affiliation(s)
- S S Meyrelles
- Cardiovascular Center, University of Iowa College of Medicine, Iowa City 52242, USA
| | | | | | | | | |
Collapse
|
44
|
Carmeliet P, Collen D. Molecular analysis of blood vessel formation and disease. THE AMERICAN JOURNAL OF PHYSIOLOGY 1997; 273:H2091-104. [PMID: 9374741 DOI: 10.1152/ajpheart.1997.273.5.h2091] [Citation(s) in RCA: 46] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Blood vessels affect the quality of life in many ways. They provide an essential nutritive function during growth and repair of tissues but, on the other hand, can become affected by disorders or trauma, resulting in bleeding, thrombosis, arterial stenosis, and atherosclerosis. Three molecular systems, the vascular endothelial growth factor (VEGF) system, the plasminogen system, and the coagulation system, have been implicated in the formation and pathobiology of blood vessels. This review focuses on the role of these systems in these processes. Recent gene-targeting studies have identified VEGF as a potent modulator of the formation of endothelial cell-lined channels. Somewhat unanticipated, the initiator of coagulation is not only involved in the control of hemostasis but also in the maturation of a muscular wall around the endothelium. With different murine models of cardiovascular disease, a pleiotropic role of the plasminogen system was elucidated in thrombosis, in arterial neointima formation after vascular wound healing and allograft transplantation, in atherosclerosis, and in the formation of atherosclerotic aneurysms. Surprisingly, tissue-type plasminogen activator is also involved in brain damage after ischemic or neurotoxic insults. The insights from these gene-targeting studies have formed the basis for designing gene therapy strategies for restenosis and thrombosis, which have been successfully tested in these knockout models.
Collapse
Affiliation(s)
- P Carmeliet
- Center for Transgene Technology and Gene Therapy, Flanders Interuniversity Institute for Biotechnology, Katholieke Universiteit Leuven, Belgium
| | | |
Collapse
|
45
|
Pili R, Chang J, Muhlhauser J, Crystal RG, Capogrossi MC, Passaniti A. Adenovirus-mediated gene transfer of fibroblast growth factor-1: angiogenesis and tumorigenicity in nude mice. Int J Cancer 1997; 73:258-63. [PMID: 9335452 DOI: 10.1002/(sici)1097-0215(19971009)73:2<258::aid-ijc16>3.0.co;2-b] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Gene transfer of angiogenic growth factors with replication-deficient recombinant adenovirus (Ad) vectors may provide a new approach to the treatment of ischemic diseases. To determine if Ad-infected cells could stimulate angiogenesis in vivo and to assess the tumorigenicity of cells infected with these vectors, NIH3T3 fibroblasts infected with Ad vectors coding for human acidic fibroblast growth factor (aFGF-1) were used in angiogenic and tumorigenic assays. Infected cells induced a strong angiogenic response in vivo, while cells infected with control virus did not. Stable 3T3 transfectants expressing the FGF-1 gene were also highly angiogenic and exhibited growth in soft agar, while Ad-infected cells did not. Ad-infected cells grew transiently in nude mice, whereas 3T3 transfectants formed large tumors which grew exponentially. Extrapolation of cell dose-response curves showed that a minimum of 1.5 x 10(4) infected cells were required for transient tumor cell growth in vivo. Ad-infected cells cultured in vitro for 30 days lost their invasive phenotype and the ability for transient cell growth in nude mice. Thus, phenotypic changes induced by Ad-mediated gene transfer of FGF-1 are transient both in vitro and in vivo, suggesting that these Ad vectors do not have tumorigenic potential. Stimulation of angiogenesis by Ad-infected cells may be useful for the evaluation of anti-angiogenic and anti-tumor agents.
Collapse
Affiliation(s)
- R Pili
- Cell Biology Unit, Laboratory of Biological Chemistry (NIH), Baltimore, MD, USA
| | | | | | | | | | | |
Collapse
|
46
|
Mitcheson JS, Hancox JC, Levi AJ. Cultured adult rabbit myocytes: effect of adding supplements to the medium, and response to isoprenaline. J Cardiovasc Electrophysiol 1997; 8:1020-30. [PMID: 9300300 DOI: 10.1111/j.1540-8167.1997.tb00626.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
INTRODUCTION The aims of this study were to investigate: (1) the effect of supplementing the culture medium on preservation of L-type calcium channel current (1Ca,L) in adult rabbit ventricular myocytes cultured for 4 days; and (2) preservation of the ICa,L response in cultured myocytes to the beta-adrenergic agonist isoprenaline. METHODS AND RESULTS Adult rabbit myocytes were cultured on laminin-pretreated glass coverslips. The basic, serum-free culture medium was supplemented with 2 mM L-carnitine, 5 mM creatine, and 5 mM taurine. Myocytes were whole cell patch-clamped, and the L-type Ca channel current was recorded selectively as Ba flux (IBa,L) via the channels. IBa,L density (i.e., IBa,L amplitude normalized to membrane capacitance) in myocytes maintained in supplemented medium did not change significantly during culture (P > 0.1). By comparison, IBa,L density in myocytes cultured in nonsupplemented medium declined by 36% after 24 hours in culture (day 1) and then recovered by the fourth day (day 4). There was no significant difference in the response to isoprenaline of acutely isolated myocytes and 4-day cultured myocytes. Isoprenaline 100 nM increased peak IBa,L by 149% +/- 32% (mean +/- SEM) in acutely isolated myocytes (n = 4 cells), and by 224% +/- 60% (n = 6 cells) and 159% +/- 24% (n = 8 cells) in day 1 and 4 cultured myocytes, respectively. CONCLUSIONS Supplemented medium improved IBa,L density in cultured myocytes. beta-Adrenergic receptors and intracellular messenger pathways appear to remain intact in adult rabbit myocytes cultured for up to 4 days.
Collapse
Affiliation(s)
- J S Mitcheson
- Department of Physiology, University of Bristol, School of Medical Sciences, United Kingdom.
| | | | | |
Collapse
|
47
|
Carmeliet P. Insights from gene-inactivation studies of the coagulation and plasminogen. ACTA ACUST UNITED AC 1997. [DOI: 10.1016/s0268-9499(97)80095-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
48
|
Nyberg-Hoffman C, Shabram P, Li W, Giroux D, Aguilar-Cordova E. Sensitivity and reproducibility in adenoviral infectious titer determination. Nat Med 1997; 3:808-11. [PMID: 9212113 DOI: 10.1038/nm0797-808] [Citation(s) in RCA: 165] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
49
|
Rust EM, Westfall MV, Samuelson LC, Metzger JM. Gene transfer into mouse embryonic stem cell-derived cardiac myocytes mediated by recombinant adenovirus. In Vitro Cell Dev Biol Anim 1997; 33:270-6. [PMID: 9156342 DOI: 10.1007/s11626-997-0046-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The main purpose of this study was to examine, for the first time, the ability of recombinant adenovirus to mediate gene transfer into cardiac myocytes derived from mouse embryonic stem (ES) cells differentiating in vitro. In addition, observations were made on the effect of adenovirus infection on cardiac myocyte differentiation and contractility in this in vitro system of cardiogenesis. ES cell cultures were infected at various times of differentiation with a recombinant adenovirus vector (AdCMVlacZ) containing the bacterial lacZ gene under the control of the cytomegalovirus (CMV) promoter. Expression of the lacZ reporter gene was determined by histochemical staining for beta-galactosidase activity. LacZ expression was not detected in undifferentiated ES cells infected with AdCMVlacZ. In contrast, infection of differentiating ES cell cultures showed increasing transgene expression with continued time in culture. Expression in ES-cell-derived cardiac myocytes was demonstrated by codetection of beta-galactosidase activity and troponin T with indirect immunofluorescence. At 24 h postinfection, approximately 27% of the cardiac myocytes were beta-galactosidase positive, and lacZ gene expression appeared to be stable for up to 21 d postinfection. Adenovirus infection had no apparent effect on the onset, extent, or duration of spontaneously contracting ES-cell-derived cardiomyocytes, indicating that cardiac differentiation and contractile function were not significantly altered in the infected cultures. The demonstration of adenovirus-mediated gene transfer into ES-cell-derived cardiac myocytes will aid studies of gene expression with this in vitro model of cardiogenesis and may facilitate future studies involving the use of these myocytes for grafting experiments in vivo.
Collapse
Affiliation(s)
- E M Rust
- Department of Physiology, University of Michigan Medical School, Ann Arbor 48109-0622, USA
| | | | | | | |
Collapse
|
50
|
Hedin U, Wahlberg E. Gene therapy and vascular disease: potential applications in vascular surgery. Eur J Vasc Endovasc Surg 1997; 13:101-11. [PMID: 9091140 DOI: 10.1016/s1078-5884(97)80004-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Advances in molecular biology have generated methods that are used to enhance diagnosis and treatment of a variety of human diseases. More recently modification of gene expression in cells by gene transfer has been introduced as a new therapeutic modality. The targeting of vascular cells with this method is appealing not only for anatomical reasons, but also because endovascular techniques provide access to the vasculature and makes site-specific delivery possible. Over the past few years, gene transfer has been widely used to explore the pathophysiology of vascular diseases in experimental models and available data suggests that this method may eventually become a therapeutic alternative for vascular disorders such as restenosis, graft failure, and critical ischaemia. In the following we discuss the methodology of gene transfer, its tentative use in vascular diseases related to vascular surgery, and the problems associated with this new technology.
Collapse
Affiliation(s)
- U Hedin
- Department of Surgery, University of Washington Medical Center, Seattle, USA.
| | | |
Collapse
|