<<

Gene Therapy (2000) 7, 2087–2093  2000 Macmillan Publishers Ltd All rights reserved 0969-7128/00 $15.00 www.nature.com/gt ACQUIRED DISEASES RESEARCH ARTICLE ␤ Direct intra-cardiomuscular transfer of 2- receptor gene augments cardiac output in cardiomyopathic hamsters

K Tomiyasu1, Y Oda1, M Nomura2, E Satoh2, S Fushiki3, J Imanishi2, M Kondo1 and O Mazda2 1First Department of Internal Medicine and 2Department of Microbiology and 3Department of Dynamic Pathology, Research Institute for Neurological Disease and Geriatrics, Kyoto Prefectural University of Medicine, Kamikyo, Kyoto, Japan

␤ ␤ In chronic heart failure, down-regulation of -adrenergic the 2-AR gene transfer. Systemic loading of isoproterenol receptor (␤-AR) occurs in cardiomyocytes, resulting in low increased the cardiac parameters more significantly on day catecholamine response and impaired cardiac function. To 2 to day 7, indicating that the adrenergic response was aug- correct the irregularity in the ␤-AR system, ␤-AR gene was mented by the genetic transduction. The same procedure transduced in vivo into failing cardiomyocytes. The Epstein– did not affect the cardiac function of normal hamsters. Immu- ␤ ␤ Barr virus (EBV)-based plasmid vector carrying human 2- nohistochemical examinations demonstrated human 2-AR AR gene was injected into the left ventricular muscle of expression in failing cardiomyocytes transduced with the Bio14.6 cardiomyopathic hamsters whose ␤-AR is down- gene. RT-PCR analysis detected mRNA for the transgene regulated in the cardiomyocytes. The echocardiographic in the heart but not in the liver, spleen, or kidney. The pro- examinations revealed that stroke volume (SV) and cardiac cedures may provide a feasible strategy for gene therapy of output (CO) were significantly elevated at 2 to 4 days after severe heart failure. Gene Therapy (2000) 7, 2087–2093.

Keywords: heart failure; ␤-; Epstein–Barr virus-based-plasmid vector; cardiomyopathic hamster; gene therapy

␤ Introduction To express a large number of 2-AR in failing cardiac muscle cells, it is necessary to find ways to conduct gen- In chronic cardiac failure, persistent excitation of the etic transduction as efficiently as possible. Previously, we sympathetic nervous system ultimately leads to down- reported that a high rate of gene transduction could be ␤ regulation of the -AR in cardiac cells, contributing to achieved in cardiomyocytes in vivo through injection of ␤ the decrease in cardiac function. If a large number of - the Epstein–Barr virus (EBV)-based plasmid.8 In the AR can be expressed in cardiomyocytes through genetic ␤ present study, we used this method to transduce the 2- transduction technique, reactivity to catecholamines may AR gene into the hearts of cardiomyopathic hamsters. be improved, enabling the possible recovery of cardiac Because direct injection of naked plasmid DNA allows 1–3 4 contractility. Milano et al established a transgenic transgene expression specifically in cardiac muscle, and ␤ mouse constitutively expressing 2-AR in the heart. They employment of the EBV-based plasmids enables highly found a significant increase in the heart rate (HR), as well efficient gene transduction, our system is considered to as improvement in the dp/dt max, an index of isochronic be highly appropriate for gene therapy of heart failure. contractility. More recently, Kawahira et al5 and Kypson 6 ␤ et al transduced the 2-AR gene ex vivo into donor hearts isolated from pressure-overloaded and normal rats, Results respectively. They found elevated LV dp/dt max in the We first estimated the transduction efficiency in vivo into heart graft heterotopically transplanted into recipient ani- cardiac muscle using our system. pSES.␤ (Figure 1) was 7 mals. Maurice et al transduced a normal rabbit heart in inoculated into the hearts of the F1b hamsters, and X-gal ␤ vivo with the 2-AR gene resulting in the elevation of LV staining was performed. The ␤-gal expression was dem- dp/dt max. Although these earlier reports strongly sug- onstrated in cardiac muscle within approximately 0.1 cm ␤ gest the feasibility of the 2-AR gene transfer in terms of from each injection site (Figure 2a). This is compatible gene therapy for heart failure, there have been no reports with our previous finding using normal rat heart.8 ␤ showing in vivo transduction of the 2-AR gene into fail- ␤ Next, the Bio14.6 hamsters were injected with pSES. 2- ing hearts, with attendant cardiac functional restoration. AR or pSES.␤ into the left ventricles, and their cardiac function was evaluated before and 2 and 4 days after the transduction. Figure 3 and Table 1 summarize the cardiac function parameters obtained by the echocardiographic Correspondence: O Mazda, Department of Microbiology, Kyoto Pre- ␤ fectural University of Medicine, Kamikyo, Kyoto 602–8566, Japan analyses. The hamsters transduced with pSES. 2-AR gene Received 11 April 2000; accepted 30 August 2000 showed 1.6-fold higher cardiac output (CO) than the ␤2-AR gene transfer into failing heart K Tomiyasu et al 2088

␤ Figure 1 Plasmids used in this study. Maps of pSES. 2-AR (left) and pSES.␤ (right)6,7 are shown. Prom, promoter; polyA, SV40 polyA additional signal.

pSES.␤-transduced animals on day 4 after the gene trans- fer (45.6 ± 21.0 ml versus 70.8 ± 21.3 ml, P Ͻ 0.05) (Figure 3, left lower panel). Cardiac response to isoproter- enol, a ␤-AR agonist, was also studied by systemic load- ␤ ing of the drug. On day 4, pSES. 2-AR-transduced group showed 1.5-, 1.3- and 1.5-fold higher value for fractional shortening (FS), stroke volume (SV) and CO, respectively, in comparison with the pSES.␤-transduced group (FS: 22.7 ± 1.5 ml versus 14.9 ± 5.7 ml, P Ͻ 0.05; SV: 0.256 ± 0.048 ml versus 0.204 ± 0.081 ml, P Ͻ 0.05; CO: 95.8 ± 20.7 ml versus 63.5 ± 31.8 ml, P Ͻ 0.05) (Figure 3, ␤ right panels). In the group given pSES. 2-AR, the FS was 1.3-fold higher than that obtained before transduction. (FS: 22.7 ± 1.5 ml versus 17.0 ± 2.9 ml, P Ͻ 0.05) (Figure 3, right upper panel). The SV and CO of the gene-trans- duced animals were elevated 1.7- and 1.8-fold on day 2, respectively, in comparison with the levels before the genetic transduction (SV: 0.296 ± 0.023 ml versus 0.170 ± 0.037 ml, P Ͻ 0.005; CO: 112.3 ± 16.7 ml versus 62.9 ± 13.3 ml, P Ͻ 0.005) (Figure 3, right middle and lower panels). The results indicate that intramyocardial ␤ injection of pSES. 2-AR elicits improvement in cardiac function, as well as augmentation of adrenergic response in the cardiomyopathic hamsters. To examine the influence of gene transduction on car- diac function of normal animals, the F1b hamsters were also tested. Compared with Bio14.6, the F1b hamsters show smaller SV and CO, due to smaller left ventricular internal dimension at end-diastole (Dd) and left ventricu- lar internal dimension at end-systole (Ds) (Tables 1 and ␤ 2). When the F1b were transduced with pSES. 2-AR or pSES.␤, no significant change was observed in the para- meters before and subsequent to genetic transduction, regardless of the plasmid injected (Table 2). Systemic administration with isoproterenol showed that the adre- ␤ nergic response in pSES. 2-AR-transduced F1b was com- parable to that in F1b given transduction with control plasmid. Next, the parameters for cardiac function were com- ␤ puted for the Bio14.6 animals that received pSES. 2-AR Figure 2 Gene transfer in vivo by intra-myocardial injection of plasmid or pSES.␤ transduction, and kinetic changes were moni- DNA. (a) Three hundred ␮g of pSES.␤ was injected into the left ventricle tored up to the 9th day following transduction. In the of F1b, and 5 days later, the animal was killed. The heart was fixed and ␮ ␤ group transduced with pSES.␤ -AR, significant elevation stained with X-gal. (b and c) Three hundred g of pSES. 2-AR (b) or 2 ␤ in the SV and CO was seen on day 2 to 7, compared with pSES. (c) was injected into the left ventricle of the Bio14.6, and 5 days later, the animal was killed. A frozen section of the heart was immuno- the levels before genetic transduction (Figure 4). ␤ ␤ stained with anti-human 2-AR antibody. The human 2-AR was demonstrated in cardio- ␤ myocytes from the pSES. 2-AR-transduced Bio14.6 by immunohistochemical staining (Figure 2b). In contrast, the hearts of pSES.␤-transduced Bio14.6 did not stain

Gene Therapy ␤2-AR gene transfer into failing heart K Tomiyasu et al 2089 there have been no reports showing in vivo genetic transduction into failing cardiomyocytes. ␤ ␤ Unlike the 2-AR transgenic mouse, the 1-AR trans- genic mouse did not show elevated cardiac contrac- 9,10 ␤ tility. Therefore, the 2-AR gene may be more suitable ␤ for gene therapy of heart failure than the 1-AR gene. The in vivo genetic transduction into heart tissue has been done using many different methods, including direct intra-myocardial injection of naked plasmid DNA11 and adenoviral vector,12 and intra-coronary infusion of adenoviral,13 HVJ-liposome14 and AAV15 vectors. We chose the direct intra-muscular injection method so that the gene expression is limited exclusively to cardiac mus- cle. Actually, we did not detect any evidence for trans- gene expression in organs other than the heart by RT- PCR (Figure 5). Another advantage of this method is sim- plicity and safety. Unlike healthy animals, the physiologi- cal data of emaciated animals like cardiomyopathic ham- sters fluctuate considerably with even a small degree of invasion, and for this reason, it is necessary to minimize the amount of invasion as much as possible. Provided that direct injection is carried out under echocardiogra- phy guidance, in vivo naked plasmid injection can be done percutaneously without thoracotomy, so that the degree of invasion with this method is very low. On the other hand, direct injection to cardiac muscle is advan- tageous over the intra-coronary approach in that the for- mer is also considered appropriate for genetic transduc- tion to cells in an ischemic lesion barely supplied with blood flow. When cardiac incompetence caused by ischemia is considered as a target disease for gene therapy, this point may become important. An impediment to naked plasmid injection is that the Figure 3 Improvement of cardiac function of gene-transduced Bio14.6 ␤ intensity of gene expression is comparatively low. To cardiomyopathic hamsters. The Bio14.6 were transduced with pSES. 2- overcome this problem, we used the EBV-based plasmid AR (solid lines, n = 7) or pSES.␤ (dotted lines, n = 6). On the indicated days after the transduction, echocardiographic examination was performed vector, which carries oriP and EBNA1 gene from EBV before (left panels) or 5 min after (right panels) percutaneous adminis- genome. The EBNA1-oriP system facilitates nuclear local- tration of 0.1 mg/kg of isoproterenol. Results are expressed as mean ± s.d. ization of the plasmid, anchorage of plasmid to nuclear (+:PϽ 0.05, ++:PϽ 0.005 in comparison with the parameters for pre- matrix, and transcriptional up-regulation. Previously, we Ͻ Ͻ ␤ transduction; *: P 0.05, **: P 0.005 in comparison between pSES. 2- have reported the effectiveness of the vectors in transfect- AR-transduced and pSES.␤-transduced groups). ing various cells of human origin both in vitro and in vivo.8,16–20 We also reported that by using the EBV-based ␤ plasmid vectors, in vitro transient expression over 10 with the anti-human 2-AR antibody that does not recog- ␤ times higher than that by using conventional plasmids nize hamster 2-AR (Figure 2c). RT-PCR experiments ␤ was observed, not only in primate cells but in various demonstrated the transgene-specific mRNA in pSES. 2- 8 AR-injected heart, but not in liver, kidney or spleen of types of rodent cells as well. Furthermore, by directly the same animal (Figure 5). The sense and antisense PCR injecting the EBV-plasmid into rat heart, higher rates of ␣ ␤ genetic transduction and expression were obtained than primers were set corresponding to SR and 2-AR ␤ by injecting conventional plasmids.8 sequences, respectively, so that endogenous 2-AR mRNA is not amplified. The results indicate that the With naked plasmid injection, genetic transduction is transgene expression was achieved exclusively in only possible in a limited portion of the heart. We cau- the heart by intra-myocardial injection of the naked tiously injected a small amount of a DNA solution into EBV-based plasmid vector. five locations of the heart, ie the front, rear, and side walls of the left ventricle, the septum, and the apex of the heart, to achieve gene expression over a comparatively wide Discussion range of cardiac muscle (Figure 2a). On the other hand, In the present study, we succeeded in augmenting car- cardiomyocytes are connected by the gap junctions, so diac function in cardiomyopathic hamsters by transfect- that adjacent cells are biochemically aligned and function ␤ ing 2-AR gene into the cardiac muscle, in an effort to cooperatively. It has been reported that cAMP, the correct irregularities in the ␤-AR system of failing cardio- second messenger mediating adrenergic stimulation, can myocytes. Administration with isoproterenol resulted in be transmitted through gap junctions.21–23 Taking advan- an even more significant improvement in cardiac func- tage of this cell-to-cell communication, it may be possible tion. The increase in CO may be ascribed to an increase to increase the contractility of cardiac cells which fail to in the SV rather than that in the HR. As far as we know, be transduced, but which are connected to neighboring

Gene Therapy ␤2-AR gene transfer into failing heart K Tomiyasu et al 2090 46 40 2.3 3.7 0.2 0.2 0.1 0.6 0.015 0.018 1.5 5.0 ± ± ± ± ± ± ± ± ± ± ± ± 4) 4) = = 41 396 40 404 7.5 37.0 3.4 60.2 0.7 4.9 0.5 4.4 0.7 3.1 0.3 2.1 0.036 0.087 0.027 0.074 12.8 34.1 11.4 29.5 ± ± ± ± ± ± ± ± ± ± ± ± 2AR (n 2AR (n

␤ ␤ 14 334 12 389 6.4 39.8 4.5 55.5 0.5 5.1 0.5 4.4 0.4 3.1 0.4 2.0 0.027 0.106 0.029 0.078 10.5 35.6 13.3 30.4 ± ± ± ± ± ± ± ± ± ± ± ± F1b F1b 56 407 31 448 2.9 34.5 5.3 53.3 0.6 4.9 0.5 4.6 0.3 3.1 0.3 2.2 0.036 0.087 0.274 0.087 13.0 35.5 11.7 39.2 ± ± ± ± ± ± ± ± ± ± ± ± 5) pSES. 5) pSES. = = 43 368 20 424 6.2 39.0 6.4 55.4 0.3 5.2 0.3 4.1 0.4 3.2 0.3 1.8 0.021 0.106 0.014 0.063 8.2 45.5 6.1 27.7 ± ± ± ± ± ± ± ± ± ± ± ± (n (n

␤ ␤ 44 323 17 389 4.7 37.6 3.6 54.8 0.2 5.0 0.3 4.3 0.3 3.2 0.3 1.9 0.010 0.095 0.011 0.071 3.2 31.0 5.3 28.0 ± ± ± ± ± ± ± ± ± ± ± ± 64 370 49 420 1.7 45.8 1.5 53.2 0.3 4.5 0.5 4.2 0.4 2.5 0.4 2.0 0.029 0.077 0.048 0.067 21.3 28.7 20.7 27.9 ± ± ± ± ± ± ± ± ± ± ± ± ) ) 7) pSES. 7) pSES. + − = = 39 308 45 372 1.9 14.8 3.3 22.7 0.5 8.2 0.3 7.8 0.4 6.9 0.5 6.1 0.04 0.228 0.023 0.256 19.3 70.8 16.7 95.8 ± ± ± ± ± ± ± ± ± ± ± ± 2AR (n 2AR (n

␤ ␤ 20 325 16 382 4.1 13.7 2.9 20 0.7 8.2 0.6 8.3 0.7 7.0 0.6 6.5 0.041 0.203 0.037 0.296 15 67.2 13.3 112.3 ± ± ± ± ± ± ± ± ± ± ± ± 73 329 66 370 3.6 13.2 5.7 17 0.4 7.7 0.5 7.4 0.4 6.7 0.7 6.2 0.053 0.156 0.081 0.17 21.0 51.6 31.8 62.9 ± ± ± ± ± ± ± ± ± ± ± ± 6) pSES. 6) pSES. = = 56 246 47 306 3.4 11.4 2.8 14.9 0.5 8.2 0.4 8.1 0.7 7.2 0.5 6.9 0.029 0.174 0.033 0.204 14.8 45.6 12.9 63.5 (n (n ± ± ± ± ± ± ± ± ± ± ± ±

␤ ␤ 38 290 29 345 3.8 12.7 3.2 18.9 0.4 8.0 0.3 7.9 0.6 7.0 0.5 6.4 0.028 0.171 0.014 0.233 12.9 49.1 6.9 79.9 ± ± ± ± ± ± ± ± ± ± ± ± s.d. s.d. ± ± Cardiac function parameters of gene-transduced animals under isoproterenol( Cardiac function parameters of gene-transduced animals under isoproterenol( bpm bpm × × Table 2 transfection pSES. transfection pSES. Table 1 HamsterDaysHR, bpm 317 0Values are mean 2 Bio 14.6 4Hamster 0DaysHR, bpm 2 358 0 4Values are mean 2 Bio 0 14.6 4 2 0 4 2 0 4 2 0 4 2 4 0 2 4 FS, % 13.7 FS, % 19.9 Dd, mm 7.8 Dd, mm 7.9 Ds, mm 6.7 Ds, mm 6.0 SV, ml 0.167 SV, ml 0.197 CO, 53.0 CO, 70.5 ml ml

Gene Therapy ␤2-AR gene transfer into failing heart K Tomiyasu et al

␤ ␤ 2091 cells that are successfully transduced with the 2-AR survival of patients. This may be ascribed to -AR down- gene. regulation in failing cardiomyocytes. Therefore, ␤-AR In our experiments, no significant change was gene transfer may be advantageous over pharmacologic observed in cardiac function of normal hamsters, while interventions. In contrast, ␤-AR blockade was revealed Kypson et al6 and Maurice et al7 reported significant elev- to be potentially useful for long-term treatment.29,30 The ation in LV-dp/dt max in normal animals given aden- concepts are compatible with the manifestations of the ␤ ␣ ovirus vector-mediated transduction of 2-AR gene. transgenic animals overexpressing the myocardial Gs ,a Because our method involved plasmid injection to car- component of ␤-AR signaling pathway.31 In cases of diac muscle, the intensity of ␤-AR expression may be chronic severe heart failure beyond conventional medical lower than that in their experiments. Alternatively, the treatment, temporary recovery of cardiac function is discrepancy may be ascribed to the different parameters mandatory. Enhancing cardiac function by a ␤-AR measured as an index of cardiac function. We analyzed increase is potentially the treatment of choice. ␤-AR gene cardiac function of animals less invasively and more therapy may be advantageous in terms of improvement physiologically using echocardiography,24 while both of quality of life in acute crisis of chronic heart failure, Kypson et al,6 Maurice et al7 and Kawahira et al5 measured under enhanced sympathetic activation. Short-term adre- LV-dp/dt max by means of cardiac catheterization. nergic stimulation may be effective in weaning from A variety of therapeutic approaches have been devised large-dose catecholamine therapy, as well as the intro- to augment the ␤-adrenergic signaling to compensate duction of ␤-AR blockade therapy in severe cases. The cardiac function in heart failure (eg administration therapy may also be applicable to temporal restoration with ,25 ,26 ,27 and of severe heart failure in patients to be treated by heart milrinone28). These therapeutic strategies actually transplantation. improve clinical symptoms during short periods follow- In conclusion, improved cardiac function was observed ␤ ing administration, but fail to significantly prolong the as a result of in vivo 2-AR genetic transduction in an animal model with failing heart. This suggests new possi- bilities in molecular treatment of severe cardiac failure in which there are obstacles to the ␤-AR system, and which are resistant to conventional medical treatment. Materials and methods Plasmids ␤ ␤ pSES. 2-AR and pSES. are plasmid vectors containing ␤ the expression units for the human 2-AR cDNA (EcoRI–

Figure 4 Kinetic change in cardiac function of gene-transduced Bio14.6 ␤ cardiomyopathic hamsters. The Bio14.6 were transduced with pSES. 2- Figure 5 Transgene is exclusively expressed in the heart. The Bio14.6 = ␤ = ␤ AR (right panels, n 7) or pSES. (left panels, n 3). On the indicated were transduced with pSES. 2-AR, and 10 days later, RNA was extracted days after the transduction, echocardiographic examination was performed from the indicated organs. RT-PCR was performed using the combinations ␣ ␤ 5 min after percutaneous administration of 0.1 mg/kg of isoproterenol. of SR -specific sense and human 2-AR gene-specific antisense primers Results are expressed as mean ± s.d. (+:PϽ 0.05 in comparison with (upper panel) or ␤-actin gene-specific sense and antisense primers pre-transduction.) (lower panel).

Gene Therapy ␤2-AR gene transfer into failing heart K Tomiyasu et al

2092 ␤ SalI 1.8 kb fragment from pTZ 2-AR kindly provided by bation with peroxidase-labeled goat anti-rabbit immuno- 32–34 ␤ ␤ Dr M Bouvier) and E. coli -gal gene, respectively. globulin (EnVision; Dako, Carpinteria, CA, USA). The 2- Both plasmids also possess the EBV oriP element, as well AR expression was visualized by treatment with DAB as the EBV nuclear antigen 1 (EBNA1) gene (Figure 1). solution (Dako).

Animals RT-PCR Bio14.6 (cardiomyopathic hamsters) and F1b (normal The organs were removed from the dead animals and golden hamsters) were purchased from Charles River rinsed with PBS. RNA was extracted with ISOGEN Laboratories (Wilmington, MA, USA). As Bio14.6 grow (Nippon Gene, Tokyo, Japan) according to the manufac- older, they show manifestations of heart failure, ␤-ARs turer’s protocol, and RT-PCR was performed as described being down-regulated in the cardiomyocytes.35 The FS of elsewhere.36 The combinations of SR␣-specific sense and the Bio14.6 were 46.8 ± 4.17 and 13.46 ± 3.78 at 29 and 45 ␤2-AR gene-specific antisense primers (5′-CCGCC weeks of age, respectively, while that of the 45-week-old TGTGGTGCCTCCTGA and 5′-TGCCCATGCCCACCA F1b was 40.78 ± 7.86. Therefore, we used 45-week-old CCCAC, respectively) or ␤-actin gene-specific sense and Bio14.6 for the experiments. antisense primers (5′-GTGCTATCCCTGTACGCCTC and 5′-AGTCCGCCTAGAAGCATTTG, respectively) were Gene transfer used (Figure 1). The animals were anesthetized by intra-peritoneal injec- tion with sodium thiobutabarbital (0.1 mg/kg). A DNA/20% sucrose solution (2 ␮g/␮l) containing a small Acknowledgements amount of carbon particles as a marker was percu- ␤ We thank Dr M Bouvier for providing the 2-AR cDNA. taneously injected into the left ventricular wall, using a This work was supported by a grant-in-aid for scientific microsyringe with a 27G needle, under guidance of the research from the Ministry of Education, Science and echocardiography (Digital Sonolayer PowerVision SSA- Culture, a grant from the Japan Heart Foundation and 380A equipped with a 7.5-MHz transducer; Toshiba, IBM Japan Research Grant, and a grant from Japan ␮ Tokyo, Japan). Approximately 40 l of the solution was Foundation of Cardiovascular Research. injected at each of five locations.

Measurement of cardiac function References The animals were anesthetized by intra-peritoneal injec- ␤ 1 Kobilka BK et al. cDNA for the human 2-adrenergic receptor: tion with sodium thiobutabarbital (0.1 mg/kg) and a protein with multiple membrane-spanning domains and enco- analyzed by blinded observers. The HR, FS, Dd and Ds ded by a gene whose chromosomal location is shared with that parameters were individually measured by the echocar- of the receptor for platelet-derived growth factor. Proc Natl Acad diographic device before or 5 min after percutaneous Sci USA 1987; 84: 46–50. administration of 0.1 mg/kg of isoproterenol. The SV and 2 Drazner MH et al. Potentiation of ␤-adrenergic signaling by CO were calculated according to following formula: adenoviral-mediated gene transfer in adult rabbit ventricular myocytes. J Clin Invest 1997; 99: 288–296. 3 3 SV = Dd − Ds 3 Akhter SA et al. Restoration of ␤-adrenergic signaling in failing CO = SV × HR cardiac ventricular myocytes via adenoviral-mediated gene trasfer. Proc Natl Acad Sci USA 1997; 94: 12100–12105. Statistical analysis 4 Milano CA et al. Enhanced myocardial function in transgenic ␤ Differences in cardiac function data were assessed by use mice overexpessing the 2-adrenergic receptor. Science 1994; 264: of a two way repeated-measures ANOVA. If a significant 582–586. 5 Kawahira Y et al. In vivo transfer of a ␤–adrenergic receptor gene F ratio was obtained, further analysis was carried out into the pressure-overloaded rat heart enhances cardiac with Fisher’s PLSD post hoc test. Differences between response to ␤-. Circulation 1998; 98: II-262–268. Ͻ ␤ groups were considered significant at P 0.05. 6 Kypson AP et al. Adenovirus-mediated gene transfer of the 2- adrenergic receptor to donor hearts enhances cardiac function. X-gal staining Gene Therapy 1999; 6: 1298–1304. The F1b were killed on day 5 following pSES.␤ transduc- 7 Maurice JP et al. Enhancement of cardiac function after adenovi- ␤ tion. The hearts were excised, cut into slices and fixed ral-mediated in vivo intracoronary 2-adrenergic receptor gene with 2% paraformaldehyde for 4 h at 4°C. Following tho- delivery. J Clin Invest 1999; 104: 21–29. 8 Tomiyasu K et al. Gene transfer in vitro and in vivo with Epstein– rough rinsing with PBS, the slices were incubated in an Barr virus-based episomal vector results in markedly high tran- X-gal staining solution (0.05% (v/v) 5-bromo-4-chloro-3- ␤ sient expression in rodent cells. Biochem Biophys Res Commun indolyl- -d-galactoside (X-gal) (Nacalai Tesque, Kyoto, 1998; 253: 733–738. Japan), 1 mm MgCl2, 150 mm NaCl, 3 mm K4[Fe(CN)6], 9 Bertin B et al. Specific atrial overexpression of G protein coupled ␤ 3mm K3[Fe(CN)6], 60 mm Na2HPO4,40mm NaH2PO4 human 1 adrenoceptors in transgenic mice. Cardiovasc Res 1993; and 0.1% Triton X-100) for 3 h at 37°C. 27: 1606–1612. 10 Bisognano JD et al. Myocardial-directed overexpression of the ␤ ␤2-AR immunohistochemistry human 1-adrenergic receptor in transgenic mice. J Mol Cell Car- The Bio14.6 were killed on day 5 after pSES.␤ -AR trans- diol 2000; 32: 817–830. 2 11 Lin H et al. Expression of recombinant genes in myocardium in duction. The hearts were excised, embedded in paraffin, vivo after direct injection of DNA. Circulation 1990; 82: 2217– and sliced to a thickness of 5 ␮m. The slices were then ␤ 2221. incubated with an appropriate dilution of anti-human 2- 12 Stratford-Pericaudet LD, Makeh I, Perricaudet M, Briand P. AR antibody (Santa Cruz Biotechnology, Santa Cruz, CA, Widespread long-term gene transfer to mouse skeletal muscles USA), followed by rinsing with PBS and further incu- and heart. J Clin Invest 1992; 90: 626–630.

Gene Therapy ␤2-AR gene transfer into failing heart K Tomiyasu et al 2093 13 Barr E et al. Efficient catheter-mediated gene transfer into the 25 Krell MJ et al. Intermittent, ambulatory dobutamine infusions in heart using replication-defective adenovirus. Gene Therapy 1994; patients with severe congestive heart failure. Am Heart J 1986; 1: 51–58. 112: 787–791. 14 Sawa Y et al. Efficient gene transfer method into whole heart 26 Lambertz H, Meyer J, Erbel R. Long-term hemodynamic effects through the coronary artery with hemagglutinating virus of of prenalterol in patients with severe congestive heart failure. Japan liposome. J Thorac Cardiovasc Surg 1997; 113: 512–519. Circulation 1984; 69: 298–305. 15 Svensson EC et al. Efficient and stable transduction of cardiomy- 27 Xamoterol in Severe Heart Failure Study Group. Xamoterol in ocytes after intramyocardial injection or intracoronary perfusion severe heart failure. Lancet 1990; 336: 1–6. with recombinant adeno-associated virus vectors. Circulation 28 Packer M et al. Effect of oral milirinone on mortality in severe 1999; 99: 201–205. heart failure. New Engl J Med 1991; 325: 1468–1475. 16 Mazda O, Satoh E, Yasutomi K, Imanishi J. Extremely efficient 29 Heilbrunn SN et al. Increased ␤-receptor density and improved gene transfection by Epstein–Barr virus vectors into lympho- hemodynamic response to catecholamine stimulation during hematopoietic cell lines. J Immunol Meth 1997; 204: 143–151. long-term therapy in heart failure from dilated car- 17 Satoh E et al. Efficient gene transduction by Epstein–Barr virus- diomyopathy. Circulation 1989; 79: 483–490. based vectors coupled with cationic liposome and HVJ-lipo- 30 Waagdstein F et al. Long-term ␤-blockade in dilated cardiomy- some. Biochem Biophys Res Commun 1997; 238: 795–799. opathy. Circulation 1989; 80: 551–563. 18 Satoh E et al. Successful transfer of adenosine deaminase (ADA) + 31 Iwase M et al. Adverse effects of chronic endogenous synpathic gene in vitro into human peripheral blood CD34 cells by trans- drive induced by cardiac Gs␣ overexpression. Circ Res 1996; 78: fecting Epstein–Barr virus (EBV)-based episomal vectors. FEBS 517–524. Lett 1998; 441: 39–42. 32 Bouvier M et al. Expression of a human cDNA encording the 19 Harada Y et al. Highly efficient suicide gene expression in hepa- ␤ -adrenergic receptor in Chinese hamster fibroblasts (CHW): tocellular carcinoma cells by Epstein-Barr virus-based plasmid 2 functionality and regulation of the expressed receptors. Mol vectors combined with polyamidoamine dendrimer. Cancer Gene Pharmacol 1987; 33: 133–139. Ther 2000; 7: 27–36. ␤ ␤ 20 Maruyama-Tabata H et al. Effective suicide gene therapy in vivo 33 Suzuki T et al. Distinct regulation of 1- and 2-adrenergic recep- by EBV-based plasmid vectors coupled with polyamidoamine tors in Chinese hamster fibroblasts. Mol Pharmacol 1991; 41: dendrimer. Gene Therapy 2000; 7: 53–60. 542–548. 21 Dermot MF, Cooper F, Mons N, Karpen JW. Adenylyl cyclases 34 Rousseau G, Manter F, Bouvier M. Distinct receptor domains and the interaction between calcium and cAMP signaling. Nat- determine subtype-specific coupling and desensitization pheno- ␤ ␤ ure (Lond) 1995; 374: 421–424. types for human 1- and 2- adrenergic receptors. Mol Pharmacol 22 De Mello WC. Impaired regulation of cell communication by ␤- 1996; 49: 752–760. adrenergic receptor activation in the failing heart. Hypertension 35 Kagiya T, Hori M, Iwai K. Alterations in cardiac [H3] nitrendip- 1996; 27: 265–268. ine binding in pathogenesis in cardiomyopathic Syrian hamsters 23 Kam Y, Kim DY, Koo SK, Joe CO. Transfer of second messengers (Bio 14.6). Jpn Circ J 1986; 50: 664. through gap junction connexin 43 channels reconstituted in lipo- 36 Hirai H et al. Use of Epstein–Barr virus-based vector/ somes. Biochim Biophys Acta 1998; 1372: 384–388. hemagglutinating virus of Japan-liposome complex vector for 24 Tanaka N et al. Transthoracic echocardiography in models of targeted gene therapy of Epstein–Barr virus-associated neo- cardiac disease in the mouse. Circulation 1996; 94: 1109–1117. plasms. Biochem Biophys Res Commun 1997; 241: 112–118.

Gene Therapy