Skip to main content

β-Adrenergic gene therapy for cardiovascular disease

Abstract

Gene therapy using in vivo recombinant adenovirus-mediated gene transfer is an effective technique that offers great potential to improve existing drug treatments for the complex cardiovascular diseases of heart failure and vascular smooth muscle intimal hyperplasia. Cardiac-specific adenovirus-mediated transfer of the carboxyl-terminus of the β-adrenergic receptor kinase (βARKct), acting as a Gβγ-β-adrenergic receptor kinase (βARK)1 inhibitor, improves basal and agonist-induced cardiac performance in both normal and failing rabbit hearts. In addition, βARKct adenovirus infection of vascular smooth muscle is capable of significantly diminishing neointimal proliferation after angioplasty. Therefore, further investigation is warranted to determine whether inhibition of βARK1 activity and sequestration of Gβγ via an adenovirus that encodes the βARKct transgene might be a useful clinical tool for the treatment of cardiovascular pathologies.

Introduction

Heart failure and hypertension are primary sources of morbidity and mortality in developed countries. There are numerous pharmaceutical agents on the market today that are used to treat these complex diseases. However, such agents are still not able to cure or to ameliorate symptoms for all patients. For this reason, new drugs and therapies are constantly being sought [1].

Gene therapy that uses in vivo recombinant adenovirus-mediated gene transfer is a powerful technique that offers great potential to improve existing drug treatments. It allows for transient expression of gene products such as peptides in targeted tissue, and therefore has the potential to be a highly specific treatment. Adenovirus-mediated gene therapy has recently come under assault because of the potentially harmful inflammatory responses to the high doses of adenovirus vectors [2]. It is likely that existing vectors will need to be improved for successful application in humans, although this is not a focus of the present commentary and readers are referred to more recent detailed discussions of viral vectors [2,3,4,5].

Gene therapy and cardiovascular disease

Several recent animal studies provide compelling evidence that gene therapy will be of great benefit as a treatment for cardiovascular diseases. For example, gene therapy using transgenes that enhance the contractile function of the heart have the potential to act as a therapeutic bridge before and during cardiac transplantation. Moreover, gene therapy may be able to ameliorate complications that arise throughout critical periods of cardiovascular disease processes, such as the remodeling that occurs after myocardial infarction or the restenosis that can occur after angioplasty or stent implantation to correct blocked arteries. Therapeutic agents aside, cardiovascular gene transfer provides an invaluable tool for examining disease-based mechanisms in animal models. Results of studies from our laboratory and from others suggest that gene therapy could become a very powerful therapeutic strategy in the very near future.

Delivery of transgene

One hurdle that must be overcome in order for gene therapy to become successful in humans is a clinically relevant, reliable, and specific method of delivery of the transgene to the cardiovascular system. Several groups have demonstrated the feasibility of delivering transgenes to the heart via direct intramyocardial injection [6,7], ex vivo perfusion [8,9,10], and finally an intracoronary approach [11,12,13,14]. Direct injection of the adenovirus into the ventricular wall has been shown to induce significant expression of transgenes with a limited amount of adenovirus [6,7]. However, there is needle damage and expression is limited to the site of injection.

Although limited in scope, ex vivo delivery systems, such as during cardiac transplantation, are clinically relevant and allow for global expression of different transgenes [8,9,10]. In order to accomplish expression of the transgene, the aortic root of the donor heart is perfused with adenoviral solution and then heterotopically transplanted into the recipient [9]. This method has been used to target robust myocardial overexpression of β2-adrenergic receptors (β2ARs), which translated into a functional effect on the transplanted heart in the form of significantly enhanced basal and agonist-induced contractility [10]. Expression of the β2AR transgene was transient, and peak expression was seen at 5 days and returned to baseline levels after 14 days [9]. This method may provide a unique opportunity for genetic manipulation of the donor organ, potentially enhancing its function during the stresses of open-heart surgery. In addition, because gene delivery is carried out in the donor heart, total body adenovirus exposure is limited in the recipient, thus making it clinically attractive for the vectors that are now available.

Another successful method of gene transfer to the heart is injection of adenovirus through the coronary arteries. Recently, investigators from two laboratories, including ours, have developed a method for global myocardial transgene delivery [12,13]. This method involves injecting adenovirus into the left ventricular cavity while the aorta or aorta and pulmonary artery are cross-clamped for several seconds, allowing perfusion of the coronary arteries via the aortic root. We have shown that in vivo myocardial gene delivery in rabbits using this left ventricular/cross-clamp delivery method of either the β2AR transgene or a gene that encodes a peptide inhibitor of the βARKct can enhance cardiac function in normal hearts as well as failing hearts [13,15]. The βARKct is comprised of the last 194 amino acids of bovine βARK1, which contains the region for binding to Gβγ (Gβγ-subunits of activated hetero-trimeric G proteins), a process that is required for βARK1 activation [15]. Thus, the βARKct inhibits the activity of βARK1 through the competitive binding of Gβγ. Global in vivo myocardial gene delivery via this left ventricular/aortic cross-clamp technique has been quite successful. It is quite invasive, however, because it requires thoracotomy; thus, less invasive methods are desirable.

Recently, it has become possible to deliver therapeutic adenoviral transgenes to the beating rabbit heart in vivo via a percutaneous intracoronary catheterization injection [14]. This technique supports transgene delivery to the area of the heart that is supplied by the catheterized artery, and may be advantageous when global transgene delivery is not required. This percutaneous coronary catheterization technique does not require entrance in the thoracic cavity, and is therefore an attractive and clinically relevant technique for use in heart failure treatment. We delivered the β2AR transgene in a ventricular-specific manner after right coronary artery or left circumflex coronary artery catheterization and adenovirus injection, and found that in vivo ventricular contractility can be enhanced 5 days after this gene treatment [14].

Beta-Adrenergic receptor signaling and heart failure

The use of βAR signaling components as gene therapy for heart failure has been the subject of debate for the past couple of years [16]. It is clear that alterations in the myocardial βAR system both precede and accompany the development of heart failure in humans [17,18]. These alterations include a 50% decrease in β1ARs, and functional uncoupling of remaining β1ARs and β2ARs in the myocardium. The latter is probably mediated by a significant upregulation in the expression and activity of βARK1 [19]. Importantly, previous work in our laboratory using novel genetically engineered mice has clearly shown that enhancing βAR signaling through overexpression of β2ARs or inhibiting βARK1 via the βARKct are novel therapeutic approaches to enhance the function of the heart [20]. In fact, this strategy has prevented the development of cardiomyopathy in murine models of heart failure [20]. Thus, data in transgenic mice coupled with our initial gene therapy results in rabbits demonstrate that enhancing myocardial β-adrenergic signaling either by overexpressing β2ARs or by inhibiting βARK1 activity may provide novel molecular ventricular assistance for failing hearts.

Beta-Adrenergic receptorsignaling and vascular intimal proliferation

Not only has gene therapy with βAR signaling components been successful for treating animal models of heart failure, but we have also shown that it can be used successfully for vascular pathologies as well [21,22]. Functional expression of transgenes including the β2AR in vascular smooth muscle has been demonstrated using catheter-mediated adenovirus delivery to de-endothelialized rat carotid arteries [22]. Total βAR density was increased in the carotids after β2AR adenovirus treatment, and this resulted in an enhancement in isoproterenol-induced vasorelaxation [22]. In humans, this could be used to improve blood flow specifically to ischemic regions, and may also lead to novel therapeutic strategies for treatment of hypertension.

Percutaneous transluminal coronary angioplasty is a method that is employed to improve cardiac blood flow through compromised vessels in a number of diseases, including unstable angina and myocardial infarction [23]. However, at the site of angioplasty there is significant injury to the arterial wall, and a major limitation to this technique is induction of the accumulation and proliferation of vascular smooth muscle cells from the tunica media to the tunica intima of the arterial wall. This proliferation leads to restenosis in 30-60% of cases within 3-6 months and is a clinical process known as intimal hyperplasia [24]. Because adenovirus-mediated transfer of a functional gene to vascular smooth muscle is feasible, we have been interested in determining whether we could prevent the smooth muscle intimal hyperplasia by directing transgene expression at the site of injury. Interestingly, it appears that, for several mitogenic agents that signal through G-protein-coupled receptors, Gβγ is a critical signaling component for mitogenesis and cell proliferation. However, this has never been documented for in vivo vascular smooth muscle intimal hyperplasia. Thus, we have utilized the βARKct transgene as an inhibitor of Gβγ in a rat carotid balloon-catheter angioplasty injury model in order to investigate the role of Gβγ in this pathologic event [21]. As detailed above, because the βARKct inhibits βARK activity through competitive Gβγ binding, it serves as a specific Gβγ inhibitor. Importantly, we found that Gβγ plays a major role in vascular smooth muscle proliferation, because targeted inhibition of Gβγ by the βARKct resulted in a significant reduction in intimal hyperplasia in this rat model of arterial restenosis [21].

Conclusion

Experimental data strongly suggest a key role for the βAR signaling system in the pathogenesis of cardiovascular disease. In heart failure, increasing βAR signaling through genetic inhibition of βARK1 or overexpression of β2AR improves cardiac performance by providing molecular ventricular assistance, and may represent a novel therapeutic approach to the management of heart failure. In addition, smooth muscle intimal hyperplasia is prevented with the expression of βARKct. Therefore, vascular pathologies are also dependent on G-protein-coupled receptor signaling. Work from our laboratory and from others suggests that adenovirus-mediated gene therapy is a powerful technique for the selective, tissue-specific expression of gene products in the cardiovascular system. To date, pharmaceutical agents are able to achieve this, and current management of both heart failure and intimal hyperplasia is not capable of preventing or completely alleviating symptoms and mortality. Therefore, we remain optimistic that adenovirus-mediated gene therapy and, more specifically, the inhibition of βARK1 activity and sequestration of Gβγ via the βARKct will be an invaluable clinical tool for the treatment of cardiovascular pathologies.

References

  1. Cohn JN, Bristow MR, Chien KR, Colucci WS, Frazier OH, Leinwand LA, Lorell BH, Moss AJ, Sonnenblick EH, Walsh RA, Mockrin SC, Reinlib L: Report of the National Heart, Lung and Blood Institute special emphasis panel on heart failure research. Circulation. 1997, 95: 766-770.

    Article  CAS  PubMed  Google Scholar 

  2. Leiden JM: Human gene therapy: the good, the bad and the ugly. Circ Res. 2000, 86: 923-925.

    Article  CAS  PubMed  Google Scholar 

  3. Nabel GJ: Development of optimized vectors for gene therapy. Proc Natl Acad Sci USA. 1999, 96: 324-326. 10.1073/pnas.96.2.324.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  4. Robbins PD, Ghivizzani SC: Viral vectors for gene therapy. Pharmacol Ther. 1998, 80: 35-47. 10.1016/S0163-7258(98)00020-5.

    Article  CAS  PubMed  Google Scholar 

  5. Searle PF, Mautner V: Adenoviral vectors: not to be sneezed at. Gene Ther. 1998, 5: 725-727. 10.1038/sj/gt/3300686.

    Article  CAS  PubMed  Google Scholar 

  6. Lin H, Parmecek MS, Morle G, Bolling S, Leiden JM: Expression of recombinant genes in myocardium in vivo after direct injection of DNA. Circulation. 1990, 82: 2217-2221.

    Article  CAS  PubMed  Google Scholar 

  7. French BA, Mazur W, Geske RS, Bolli R: Direct in vivo gene transfer into porcine myocardium using replication-deficient adenoviral vectors. Circ. 1994, 90: 2414-2424.

    Article  CAS  Google Scholar 

  8. Lee J, Laks H, Drinkwater DC, Blitz A, Lam L, Shiraishi Y, Chang P, Drake TA, Ardehali A: Cardiac gene transfer by intracoronary infusion of adenovirus vector-mediated reporter gene in the transplanted mouse heart. J Thorac Cardiovasc Surg. 1996, 111: 246-252.

    Article  CAS  PubMed  Google Scholar 

  9. Kypson AP, Peppel K, Akhter SA, Lilly E, Glower DD, Lefkowitz RJ, Koch WJ: Ex-vivo adenoviral-mediated gene transfer to the transplanted adult rat heart. J Thorac Cardiovasc Surg. 1998, 115: 623-630.

    Article  CAS  PubMed  Google Scholar 

  10. Kypson A, Hendrickson S, Akhter S, Wilson K, McDonald P, Lilly R, Dolber P, Glower D, Lefkowitz R, Koch W: Adenovirus-mediated gene transfer of the β2-adrenergic receptor to donor hearts enhances cardiac function. Gene Ther. 1999, 6: 1298-1304. 10.1038/sj.gt.3300940.

    Article  CAS  PubMed  Google Scholar 

  11. Barr E, Carroll J, Kalynych AM, Tripathy SK, Kozarsjy K, Wilson JM, Leiden JM: Efficient catheter mediated gene transfer into the heart using replication-defective adenovirus. Gene Ther. 1994, 1: 51-58.

    CAS  PubMed  Google Scholar 

  12. Hajjar RJ, Schmidt U, Matsui T, Guerrero JL, Lee K-H, Gwathmey JK, Dec GW, Semigran MJ, Rosenzweig A: Modulation of ventricular function through gene transfer in vivo. Proc Natl Acad Sci USA. 1998, 95: 5251-5256. 10.1073/pnas.95.9.5251.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  13. Maurice JP, Hata JA, Shah AS, White DC, McDonald PH, Dolber PC, Wilson KH, Lefkowitz RJ, Glower DD, Koch WJ: Enhancement of cardiac function after adenoviral-mediated in vivo intracoronary Beta2-adrenergic receptor gene delivery. J Clin Invest . 1999, 104: 21-29.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  14. Shah AS, Lilly RE, Kypson AP, Tai O, Hata JA, Pippen A, Silvestry SC, Lefkowitz RJ, Glower DD, Koch WJ: Intracoronary adenovirus-mediated delivery and overexpression of the Beta2-adrenergic receptor in the heart. Prospects for molecular ventricular assistance. Circulation. 2000, 101: 408-414.

    Article  CAS  PubMed  Google Scholar 

  15. White DC, Hata JA, Shah AS, Glower DD, Lefkowitz RJ, Koch WJ: Preservation of myocardial Beta-adrenergic receptor signaling delays the development of heart failure after myocardial infarction. Proc Natl Acad Sci USA. 2000, 97: 5428-5433. 10.1073/pnas.090091197.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  16. Lefkowitz RJ, Rockman HA, Koch WJ: Catecholamines, cardiac Beta-adrenergic receptors, and heart failure. Circulation. 2000, 101: 1634-1637.

    Article  CAS  PubMed  Google Scholar 

  17. Bristow MR, Ginsburg R, Minobe W, Cubicciotti RS, Sageman WS, Lurie K, Billingham ME, Harrison DC, Stinson EB: Decreased catecholamine sensitivity and Beta-adrenergic receptor density in failing human hearts. N Engl J Med. 1982, 307: 205-211.

    Article  CAS  PubMed  Google Scholar 

  18. Brodde OE: Beta-adrenoceptors in cardiac disease. Pharmacol Ther. 1993, 60: 405-430. 10.1016/0163-7258(93)90030-H.

    Article  CAS  PubMed  Google Scholar 

  19. Ungerer M, Bohm M, Elce JS, Erdmann E, Lohse MJ: Altered expression of beta-adrenergic receptor kinase and beta 1-adrenergic receptors in the failing human heart. Circulation. 1993, 87: 454-463.

    Article  CAS  PubMed  Google Scholar 

  20. Koch WJ, Lefkowitz RJ, Rockman HA: Functional consequences of altering myocardial adrenergic receptor signaling. Annu Rev Physiol . 2000, 62: 237-260. 10.1146/annurev.physiol.62.1.237.

    Article  CAS  PubMed  Google Scholar 

  21. Iaccarino G, Smithwick LA, Lefkowitz RJ, Koch WJ: Targeting GBetaGamma signaling in arterial vascular smooth muscle proliferation: a novel strategy to limit restenosis. Proc Natl Acad Sci USA. 1999, 96: 3945-3950. 10.1073/pnas.96.7.3945.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  22. Gaballa MA, Peppel K, Lefkowitz RJ, Aguirre M, Dolber PC, Pennock GD, Koch WJ, Goldman S: Enhanced vasorelaxation by overexpression of Beta2-adrenergic receptors in larger arteries. J Mol Cell Cardiol. 1998, 30: 1037-1045. 10.1006/jmcc.1998.0668.

    Article  CAS  PubMed  Google Scholar 

  23. DeFeyter PJ, Serruys PW: In: Textbook of Interventional Cardiology. Edited by Topol EJ. Philadelphia: Saunders;. 1994, 274-291.

    Google Scholar 

  24. McBride W, Lange RA, Hillis LD: Restenosis after successful coronary angioplasty. Pathophysiology and prevention. N Engl J Med . 1988, 318: 1734-1737.

    Article  CAS  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Walter J Koch.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Eckhart, A.D., Koch, W.J. β-Adrenergic gene therapy for cardiovascular disease. Trials 1, 131 (2000). https://0-doi-org.brum.beds.ac.uk/10.1186/cvm-1-3-131

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • DOI: https://0-doi-org.brum.beds.ac.uk/10.1186/cvm-1-3-131

Keywords