Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2014 Apr 1.
Published in final edited form as: Transl Res. 2012 Dec 20;161(4):313–320. doi: 10.1016/j.trsl.2012.11.004

Gene Transfer for Congestive Heart Failure: Update 2013

Tong Tang 1, H Kirk Hammond 1
PMCID: PMC3602385  NIHMSID: NIHMS432598  PMID: 23261978

Abstract

Congestive heart failure is a major cause of morbidity and mortality with increasing social and economic costs. There have been no new high impact therapeutic agents for this devastating disease for more than a decade. However, many pivotal regulators of cardiac function have been identified using cardiac-directed transgene expression and gene deletion in preclinical studies. Some of these increase function of the failing heart. Altering the expression of these pivotal regulators using gene transfer is now either being tested in clinical gene transfer trials, or soon will be. In this review, we summarize recent progress in cardiac gene transfer for clinical congestive heart failure.

Keywords: Gene Transfer, Adenovirus, AAV, LV Contractility, Ca2+ Handling, β-adrenergic receptor

INTRODUCTION

Congestive heart failure (CHF) is a condition in which the heart cannot pump enough blood to meet the body's needs. It is a leading cause of morbidity and mortality in the world. In the US, there are approximately 300,000 deaths every year which are due to CHF.1 There are >23 million CHF patients worldwide, and this number is projected to double by 2030. Even with optimal pharmacological and device therapy, the outcome for subjects with CHF remains poor with 50% mortality within 4–5 years. Heart transplantation has an 80% 5-year survival, but is a solution for only 2,500 patients annually in the US.

This unmet medical need demands more effective options for patients with CHF. Stem cell delivery shows some promise in treating acute myocardial infarction;2 however, it has not been shown to be effective in randomized clinical trials. Gene transfer is simpler and easier to apply clinically than is stem cell delivery, and two recent clinical gene transfer trials have been initiated. Here, we first review general considerations for selection of vector, transgene, and delivery methods for cardiac gene transfer, and summarize recent progress in cardiac gene transfer for clinical CHF.

GENERAL CONSIDERATIONS FOR CARDIAC GENE TRANSFER

The chief impediment to successful CHF gene therapy is in obtaining sufficient cardiac transgene expression to provide a therapeutic effect. There are three requirements that must be met for successful CHF gene therapy: (1) a therapeutic transgene that can increase function of the failing heart; (2) a suitable vector with an appropriate promoter that will ensure long-term and sufficient transgene expression with minimal toxicity; and (3) a delivery method that safely provides effective cardiac gene transfer and yet is easy to deploy.

Cardiac myocyte-targeted transgene expression and gene deletion studies in mice have identified pivotal regulators of cardiac function that are deficient or dysfunctional in CHF. Many of these proteins are suited to gene transfer, providing new potential treatments for CHF. New virus vectors have been engineered and novel delivery methods have been tested to enable long-term and higher level expression. Virus vectors encoding a variety of therapeutic transgenes appear to increase function of the failing heart in preclinical studies,37 and thus a focus of this review. Indeed, clinical CHF trials using intracoronary delivery of an adeno-associated virus (AAV) vector encoding SERCA2a (ClinicalTrials.gov NCT01643330) and adenovirus encoding adenylyl cyclase 6 (AC6) (ClinicalTrials.gov NCT00787059) are currently in progress (Table 1). Other transgenes including βARKct and S100A1, which have shown efficacy in preclinical studies, may advance to clinical trials soon.8

Table 1.

Clinical Trials Using Cardiac Gene Transfer for CHF

Transgene SDF-1 SDF-1 SERCA2a SERCA2a AC6
Trial Identifier NCT01082094 NCT01643590 NCT00454818 NCT01643330 NCT00787059
Phase 1 2 1 and 2 2 1/2
Vector plasmid plasmid AAV1 AAV1 Adenovirus
Delivery Method Endomycardial injection Endomycardial injection Intracoronary injection Intracoronary injection Intracoronary injection
Enrollment 17 90 39 200 72
Sample Size 5 mg dose: 4
15 mg dose: 6
30 mg dose: 7
Placebo: 30
15 mg dose: 30
30 mg dose: 30
Placebo: 14
Low dose: 8
Middle dose: 8
High dose: 9
Placebo: 100
Treatment: 100
Placebo: 18
Treatment: 54 in 6 doses
Design Open label Randomized
Double-blind
Placebo-controlled
Randomized
Double-blind
Placebo-controlled
Randomized
Double-blind
Placebo-controlled
Randomized
Double-blind
Placebo-controlled
Status Completed Recruiting Completed Recruiting Recruiting
Outcome Apparently safe;
Trends in improving symptoms and functions
N/A Apparently safe;
Trends in improving symptoms and functions
N/A N/A

VECTORS AND METHODS FOR CARDIAC GENE TRANSFER

Plasmid Vectors

Plasmid vectors were used in previous gene transfer trials in angiogenesis, and this approach is being used in a current gene transfer trial in subjects with symptomatic CHF. The gene, stromal cell-derived factor 1 (SDF-1), is injected into the LV wall using a catheter-based system. Results from an open-label Phase 1 study (ClinicalTrials.gov NCT01082094; Table 1),9 supported initiation of a randomized, double-blind, placebo-controlled Phase 2 study (ClinicalTrials.gov NCT01643590). Results of the randomized trial are not available.

Virus Vectors

Adenovirus and AAV are the most commonly used vectors for cardiac gene transfer. E1,E3-deleted adenovirus provides reasonable gene transfer efficiency,10 particularly when used with mechanical or pharmaceutical adjuvants following intracoronary delivery. Newer generation adenovirus, including so-called gutless adenovirus that are engineered by deleting all regions encoding virus proteins, may have lower immunogenicity than previous vectors.

AAV, with an insert capacity <5 kb, provides potential long-term expression and, like adenovirus, is not associated with risk of insertional mutagenesis. Persistent transgene expression has been shown in rodents and larger mammals years after a single injection of AAV.11 Although recent clinical trials have found that some AAV serotypes incite immune responses after intramusclar injection,12, 13 other AAV vectors (AAV5, 6, 8 and 9) do not appear to have similar problems in non-human primates.14 Previous exposure to AAV, with subsequent generation of neutralizing antibodies, impairs the effectiveness of AAV vectors in cardiac gene transfer. Pre-existing anti-AAV8 antibodies are present in 19% of human subjects. AAV1 and AAV2 have a 50–59% prevalence of neutralizing antibodies, making these serotypes not useful in clinical applications.15

Self-complementary AAV vectors (scAAV) may provide more rapid and perhaps higher transgene expression than their single stranded (ssAAV) analogs.15 Transgene expression using ssAAV vectors is delayed four weeks until the complementary DNA strand is synthesized. By encoding for the complementary DNA strand within the vector, scAAV (insert capacity 3.3 kb), enables transgene expression in two weeks.15

Promoters

The CMV (cytomegalovirus) promoter is widely used for cardiac gene transfer. It provides strong transgene expression in cardiac myocytes. However, the CMV promoter is susceptible to methylation and subsequent inactivation in the liver and skeletal muscle.16, 17 RSV (Rous sarcoma virus), CBA (chicken β-actin), and EF1α (elongation factor 1α) provide less robust transgene expression in cardiac myocytes, but are less susceptible to methylation. None of these promoters provide cardiac-specific transgene expression.

The α-MHC (α-myosin heavy chain) promoter is used for cardiac-directed transgene expression in transgenic mice.18 However, it provides less robust transgene expression in virus vectors. More relevant, its size (~5.5 kb) is over the packaging capacity of AAV and prevents its use in the AAV-mediated cardiac gene transfer. A 2.1 kb fragment of the MLC-2v (myosin light chain 2v) promoter may be a suitable option.19 It is intriguing that a 418 bp fragment of chicken cardiac troponin T (cTnT) promoter provides 100-fold more transgene expression in the heart than liver after AAV-mediated gene transfer, although transgene expression is lower than that provided by CMV and CBA promoters.20

Gene Delivery Methods

Delivery methods are often determined by vector selection. For example, intravascular delivery is not suitable for lentivirus, a vector that is unable to cross the capillary endothelium.21 Direct intramyocardial injection appears to be the best delivery route for lentivirus for cardiac gene transfer. However, intramusclular injection provides transgene expression limited to the area adjacent to the needle tract. Direct intramyocardial injection is less efficient than intravascular delivery of adenovirus or AAV.

There are three effective delivery methods for cardiac gene transfer: direct intracoronary, indirect intracoronary (indirect IC), and IV.22, 23 In mice, the coronary arteries are too small for direct intracoronary delivery. Indirect IC delivery, instead, has been used to deliver virus vectors to the LV in mice and rats. In this procedure, the aorta and pulmonary arteries are cross-clamped and virus vectors are delivered into the LV chamber. Continued LV contraction then forces the vector into the coronary arteries. This method is used to deliver adenovirus and AAV vectors in rodents. Since vector exposure time promotes gene transfer, hypothermia is used to prolong dwell time but preclude brain injury. Pharmacological agents, such as histamine24, 25, serotonin,26 nitroprusside27, 28, sildenafil29, and substance P30, are also administrated to increase adenovirus-mediated gene transfer efficiency.31 IV delivery of AAV vectors (especially AAV6, AAV8, and AAV9) has been used with success in cardiac gene transfer,6, 8, 32 although indirect IC delivery provides superior cardiac gene transfer efficiency compared with IV delivery, regardless of AAV serotype.33

Regulated Transgene Expression

Long-term expression vectors require, for safety in clinical trials, the ability to turn off transgene expression in the event that untoward effects develop. Regulated expression also enables the flexibility of intermittent rather than constant transgene expression. There are four regulated expression systems currently available: ecdysone, tamoxifen, tetracycline, rapamycin.3437 The size of the ecdysone system requires a two-vector strategy and tamoxifen presents difficult to resolve issues with toxicity. Tetracycline and rapamycin regulation systems (Table 2) have been tested in large animal models.5, 11, 3842

Table 2.

Tetracycline vs Rapamycin Regulation

Feature Tetracycline Rapamycin
Activator Doxycycline AP22594
Basal Expression (“leak”) Very low/none None
Linear Dose-Response Yes Yes
Activator Side-effects Low (avoid in pregnancy) Immunosuppressant
Bacteria/Virus Proteins Yes No
Used in Clinical Trials Not yet Not Yet

AP22594, oral rapamycin analog with 1/100th immune suppression vs rapamycin.

Tetracycline-Regulated Expression

The tet-regulation system has been extensively studied.16 Unlike previous rtTA constructs, newer rtTA variants, such as rtTA2S-M2, provide robust tet-dependent expression with no basal activity (ie. no “leak”) and 10-fold higher sensitivity to tetracycline (maximum transgene expression activation at 0.1 µg/ml).43 A single daily dose of doxycycline of 10–20 mg may suffice for complete activation of transgene expression in human subjects.44 Doses of 200 mg/d are well tolerated by patients using oral doxycycline chronically for acne and chronic infections.44, 45 Tetracyclines may attenuate matrix metalloproteinase (MMP) activity and affect LV remodeling when administered in the first few days after MI.41 In clinical settings, tetracycline should be avoided in the acute phase of MI.

Immune responses to components of the rtTA system did not occur when AAV4.tet and AAV5.tet gene transfer (intraretinal) were used in non-human primates,38, 44 where tetracycline-dependent transgene expression persisted for the 2.5 year duration of the study. We do not see inflammation in mouse hearts expressing high levels of rtTA,41, 42, 45 or in rats after AAV5-mediated regulated expression of IGF-I using the rtTA2S-M2 regulation element.46 It appears that intramuscular delivery of AAV in nonhuman primates, unlike intraretinal or vascular delivery, does lead to attenuation of regulated expression, owing to immune responses to the bacterial and virus components of the transactivator fusion protein.47 The rapamycin-regulation system, which does not possess bacterial or virus proteins, and is not associated with provocation of the immune response,11 may be a suitable alternative.

Rapamycin-Regulated Expression

In the rapamycin regulation system, transgene expression is triggered by nanomolar concentrations of rapamycin or a rapamycin analog, which is dose-dependent and reversible.37 Rapamycin is used clinically to suppress immune response, forestalls deleterious effects of aging in mice40 and inhibits glioblastoma multiforme48 by blocking the mammalian target of rapamycin (mTOR) signaling pathway.49 The oral rapamycin analog AP22594, which activates transgene expression as effectively as rapamycin, exhibits minimal immune suppression, and does not inhibit mTOR.11, 4951 Additional preclinical studies directly comparing tetracycline-regulated and rapamycin-regulated expression will be required before using these systems in clinical trials.

Alternative Methods for Cardiac Gene Transfer

Paracrine-based gene transfer is an alternate to cardiac-targeted gene transfer and may be applicable for CHF and other cardiovascular diseases. A prerequisite for this approach is the selection of a transgene that has cardiac effects after being released to the circulation from a distant site. We have tested this concept using skeletal muscle injection of AAV5 encoding IGF-I (insulin growth factor I) under tet regulation (AAV5.IGFI-tet).46 In this study, AAV5.IGFI-tet was injected in the anterior tibialis muscle in rats with severe CHF induced by myocardial infarction. Activation of IGF-I expression by addition of doxycycline to the drinking water increased serum IGF-I levels and improved function of the failing heart. This new approach enables transgene expression at a remote site and circumvents the problem of attaining high yield cardiac gene transfer.

CANDIDATES FOR CHF GENE TRANSFER

CHF is associated with several cell signaling pathways that are dysfunctional. Consequently, several potential therapeutic targets have been identified. We will summarize here only those strategies that have proven to be effective in preclinical studies and have advanced or may soon advance to clinical trials for CHF. It is not surprising that these potential targets influence β-adrenergic receptor (βAR) and Ca2+ signaling.

βAR Signaling

A hallmark of clinical CHF is impaired left ventricular (LV) βAR signaling.52 The molecular basis for impaired βAR signaling includes decreased βAR density, βAR desensitization, uncoupling of βAR and Gαs, deficits in adenylyl cyclase (AC) expression, and subsequent defective cAMP production.53 Clinical use of βAR antagonists (beta-blockers) reduce symptoms and prolong life somewhat in CHF.54 Correcting impaired βAR signaling safely has been a focus for CHF research for many years, and presents considerable challenges.

βARKct

GRK2(G-protein-coupled receptor kinase 2) is a protein kinase that phosphorylates the βAR.53 This phosphorylation promotes βAR binding to β-arrestin, which in turn promotes Gαs uncoupling and attenuation of βAR signaling. β-arrestin binding also leads to β1AR internalization. The human failing heart is associated with increased expression and activity of GRK2.55 Conversely, in preclinical studies, deletion of GRK2 increases survival, attenuates LV remodeling, and reduces the extent of CHF after myocardial infarction.56

βARKct (C-terminal domain of GRK2) was engineered to block GRK2 membrane translocation and activation. Expression of βARKct after virus-mediated gene transfer increases βAR density, cAMP production, and LV contractile function in myocardial infarction-induced CHF in rats, rabbits, and pigs.7, 57, 58 βARKct expression in cardiac myocytes from failing human hearts also increases cAMP production, cell shortening, and relaxation.59 Importantly, βARKct may work addictively with βAR antagonists.58, 60

AC6

AC is the effector molecule that links βAR stimulation with cAMP production.6163 In the heart, it plays a pivotal role in LV contractile function and relaxation in response to βAR stimulation.24, 43, 64, 65 The failing heart is associated with decreased expression and activity of AC6, a major AC isoform in cardiac myocytes.52, 6668 Cardiac-directed expression of AC6 in a genetic animal model of CHF increases impaired LV function and prolongs life.69, 70 Associated with these beneficial effects in the failing heart are increased cAMP generating capacity in response to βAR stimulation, normalized PKA activity, increased phospholamban phosphorylation, and increased sarcoplasmic reticulum (SR) Ca2+ uptake.70, 71 AC6 expression is not associated with increased heart rate or development of arrhythmias in the failing heart.72 Increased LV AC6 expression is not associated with changes in contents of βAR, Gαs, or Gαi2 have been observed.64 Deletion of AC6 is associated with reduced SR Ca2+ uptake and decreased LV function in response to βAR stimulation.73 In addition, AC6 expression normalizes prolonged action potential duration and attenuates ventricular arrhythmias.74

AC6 expression also has a pronounced favorable effect on cardiovascular function in CHF induced by myocardial infarction. Although AC6 has no effect on infarct size, it prevents deleterious LV remodeling, and reduces mortality in acute myocardial infarction.75 When AC6 expression is activated 5 weeks after myocardial infarction, at which time severe CHF is evident, both LV systolic function and diastolic function are increased.5 Intracoronary delivery of adenovirus encoding AC6 increases LV function in the failing pig heart.27 A clinical trial of AC6 gene transfer for CHF is in progress (ClinicalTrials.gov NCT00787059). AC6 expression increases LV Ca2+ handling and LV function in aged mice suggest a potential role in elderly subjects with CHF with preserved ejection fraction.76

Ca2+ Handling

Calcium plays a crucial role in controlling LV contraction and relaxation. During every heartbeat, Ca2+ is taken up and then released from SR. The failing heart is characterized by defective excitation-contraction coupling (E-C coupling) and dysfunctional SR Ca2+ uptake and release.77

SERCA2a

SERCA2a (SR Ca2+-ATPase 2a) is the Ca2+ pump responsible for cardiac SR Ca2+ uptake. CHF is associated with abnormal SERCA2a expression and activity.78, 79 Cardiac-directed SERCA2a expression increases SR Ca2+ uptake and LV contractile function and relaxation.80, 81 Homogenous SERCA2a deletion is lethal. Mice with deletion of only one SERCA2a allele show decreased SR Ca2+ uptake and are more prone to CHF after pressure overload.82, 83 These data suggest a role of SERCA2a in mediating Ca2+ handling and LV function.

SERCA2a gene transfer increases contractile function in vitro in cardiac myocytes isolated from CHF patients.84 In pressure-overloaded hearts, expression of SERCA2a by adenovirus-mediated gene transfer increases contractile function and relaxation in rats.85, 86 SERCA2a gene transfer also increases LV function in aged hearts.87 In volume-overloaded pigs, AAV-mediated SERCA2a gene transfer attenuated LV dysfunction and remodeling.3 SERCA2a gene transfer shows similar beneficial effects in sheep with pacing-induced CHF.88 A clinical trial of SERCA2a gene transfer with 39 CHF patients was conducted, which indicated that intracoronary delivery of AAV1.SERCA2a was safe. The trial was insufficiently powered to determine efficacy although some indications of improvement were reported. A larger clinical trial of SERCA2a gene transfer is in progress (ClinicalTrials.gov NCT01643330).

S100A1

S100A1, a family member of EF-hand Ca2+–binding proteins, is expressed in cardiac myocytes.89 The subcellular locations of S100A1 include SR, mitochondria, and sarcomere. S100A1 can bind to RyR2, the SR Ca2+ release channel, and to SERCA2a, the SR Ca2+ uptake pump,90, 91 suggesting that S100A1 is not solely a Ca2+-binding protein but also a regulator for Ca2+ homeostasis in cardiac myocytes. There is evidence that ischemic cardiomyopathy is associated with decreased S100A1 expression.92

Cardiac-directed S100A1 expression is associated with improved Ca2+ handling, decreased deleterious LV remodeling, and reduced mortality after myocardial infarction in mice.93 AAV-mediated gene transfer of S100A1 in CHF increases Ca2+-transients and LV function.94 S100A1 gene transfer also improves Ca2+ handling and contractile function in vitro in cardiac myocytes isolated from human failing heart.95 Recent data demonstrate that intracoronary delivery of AAV9 encoding S100A1 normalizes SR Ca2+ handling, attenuates LV remodeling, and increases contractile function.6

Although βARKct, AC6, SERCA2a, and S100A1 likely operate via effects on βAR and Ca2+ signaling, favorable cardiac effects may be mediated by additional mechanisms. For example, both βARKct and AC6 expression activate Akt – a kinase that promotes cell survival.96, 97 S100A1 inhibits 2-deoxyglucose and oxidative stress-induced apoptosis in neonatal cardiac myocytes in vitro.98 SERCA2a and S100A1 appear to bind to eNOS in endothelial cells, suggesting a role in modulating production of NO – a pivotal molecule for blood flow regulation.99, 100 Further exploration of the underlying mechanisms may help improve the efficacy of these CHF therapy candidates.

CONCLUSION

Preclinical studies have identified potential therapeutic genes for treatment of CHF. SERCA2a and AC6 have advanced to clinical gene transfer trials. The optimization of virus vectors, regulated expression systems, gene delivery methods, and identification of new therapeutic candidates will move gene transfer for CHF to more importance in the next 10 years.

ACKNOWLEDGEMENTS

This work was supported by a Grant-in-Aid from American Heart Association (11GRNT7610059 to TT), grants from the National Institutes of Health (5P01HL066941, HL081741, HL088426 to HKH), and a Merit Review Award from the Department of Veterans Affairs (I01BX001515 to HKH).

Footnotes

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

Dr. Hammond is founder, consultant, and equity holder in Renova Therapeutics, which was not involved in any manner with the studies reviewed. Dr. Tang has no conflict of interest.

AUTHOR CONTRIBUTIONS

TT and HKH wrote and approved the final draft of the manuscript.

REFERENCES

  • 1.Roger VL, Go AS, Lloyd-Jones DM, Benjamin EJ, Berry JD, Borden WB, Bravata DM, Dai S, Ford ES, Fox CS, Fullerton HJ, Gillespie C, Hailpern SM, Heit JA, Howard VJ, Kissela BM, Kittner SJ, Lackland DT, Lichtman JH, Lisabeth LD, Makuc DM, Marcus GM, Marelli A, Matchar DB, Moy CS, Mozaffarian D, Mussolino ME, Nichol G, Paynter NP, Soliman EZ, Sorlie PD, Sotoodehnia N, Turan TN, Virani SS, Wong ND, Woo D, Turner MB. Executive summary: Heart disease and stroke statistics--2012 update: A report from the american heart association. Circulation. 2012;125:188–197. doi: 10.1161/CIR.0b013e3182456d46. [DOI] [PubMed] [Google Scholar]
  • 2.Ptaszek LM, Mansour M, Ruskin JN, Chien KR. Towards regenerative therapy for cardiac disease. Lancet. 2012;379:933–942. doi: 10.1016/S0140-6736(12)60075-0. [DOI] [PubMed] [Google Scholar]
  • 3.Kawase Y, Ly HQ, Prunier F, Lebeche D, Shi Y, Jin H, Hadri L, Yoneyama R, Hoshino K, Takewa Y, Sakata S, Peluso R, Zsebo K, Gwathmey JK, Tardif JC, Tanguay JF, Hajjar RJ. Reversal of cardiac dysfunction after long-term expression of serca2a by gene transfer in a pre-clinical model of heart failure. J Am Coll Cardiol. 2008;51:1112–1119. doi: 10.1016/j.jacc.2007.12.014. [DOI] [PubMed] [Google Scholar]
  • 4.Kaye DM, Preovolos A, Marshall T, Byrne M, Hoshijima M, Hajjar R, Mariani JA, Pepe S, Chien KR, Power JM. Percutaneous cardiac recirculation-mediated gene transfer of an inhibitory phospholamban peptide reverses advanced heart failure in large animals. J Am Coll Cardiol. 2007;50:253–260. doi: 10.1016/j.jacc.2007.03.047. [DOI] [PubMed] [Google Scholar]
  • 5.Lai NC, Tang T, Gao MH, Saito M, Takahashi T, Roth DM, Hammond HK. Activation of cardiac adenylyl cyclase expression increases function of the failing ischemic heart in mice. J Am Coll Cardiol. 2008;51:1490–1497. doi: 10.1016/j.jacc.2008.01.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Pleger ST, Shan C, Ksienzyk J, Bekeredjian R, Boekstegers P, Hinkel R, Schinkel S, Leuchs B, Ludwig J, Qiu G, Weber C, Raake P, Koch WJ, Katus HA, Muller OJ, Most P. Cardiac aav9-s100a1 gene therapy rescues post-ischemic heart failure in a preclinical large animal model. Sci Transl Med. 2011;3 doi: 10.1126/scitranslmed.3002097. 92ra64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Raake PW, Schlegel P, Ksienzyk J, Reinkober J, Barthelmes J, Schinkel S, Pleger S, Mier W, Haberkorn U, Koch WJ, Katus HA, Most P, Muller OJ. Aav6.Betaarkct cardiac gene therapy ameliorates cardiac function and normalizes the catecholaminergic axis in a clinically relevant large animal heart failure model. Eur Heart J. 2012 doi: 10.1093/eurheartj/ehr447. in press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Raake PW, Tscheschner H, Reinkober J, Ritterhoff J, Katus HA, Koch WJ, Most P. Gene therapy targets in heart failure: The path to translation. Clin Pharmacol Ther. 2011;90:542–553. doi: 10.1038/clpt.2011.148. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Penn MS, Pastore J, Rouy D, Schaer G, Farr MJ, Sherman W, Mendelsohn F, Losordo D. Initial results from a first-in-man study delivering non-viral gene therapy jvs-100 to treat ischemic heart failure. Mol Ther. 2011;19(Supple 1):S40. [Google Scholar]
  • 10.Alba R, Bosch A, Chillon M. Gutless adenovirus: Last-generation adenovirus for gene therapy. Gene Ther. 2005;12(Suppl):S18–S27. doi: 10.1038/sj.gt.3302612. [DOI] [PubMed] [Google Scholar]
  • 11.Rivera VM, Gao GP, Grant RL, Schnell MA, Zoltick PW, Rozamus LW, Clackson T, Wilson JM. Long-term pharmacologically regulated expression of erythropoietin in primates following aav-mediated gene transfer. Blood. 2005;105:1424–1430. doi: 10.1182/blood-2004-06-2501. [DOI] [PubMed] [Google Scholar]
  • 12.Mingozzi F, Meulenberg JJ, Hui DJ, Basner-Tschakarjan E, Hasbrouck NC, Edmonson SA, Hutnick NA, Betts MR, Kastelein JJ, Stroes ES, High KA. Aav-1-mediated gene transfer to skeletal muscle in humans results in dose-dependent activation of capsid-specific t cells. Blood. 2009;114:2077–2086. doi: 10.1182/blood-2008-07-167510. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Manno CS, Pierce GF, Arruda VR, Glader B, Ragni M, Rasko JJ, Ozelo MC, Hoots K, Blatt P, Konkle B, Dake M, Kaye R, Razavi M, Zajko A, Zehnder J, Rustagi PK, Nakai H, Chew A, Leonard D, Wright JF, Lessard RR, Sommer JM, Tigges M, Sabatino D, Luk A, Jiang H, Mingozzi F, Couto L, Ertl HC, High KA, Kay MA. Successful transduction of liver in hemophilia by aav-factor ix and limitations imposed by the host immune response. Nat Med. 2006;12:342–347. doi: 10.1038/nm1358. [DOI] [PubMed] [Google Scholar]
  • 14.Hildinger M, Auricchio A, Gao G, Wang L, Chirmule N, Wilson JM. Hybrid vectors based on adeno-associated virus serotypes 2 and 5 for muscle-directed gene transfer. J Virol. 2001;75:6199–6203. doi: 10.1128/JVI.75.13.6199-6203.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Wang Z, Ma HI, Li J, Sun L, Zhang J, Xiao X. Rapid and highly efficient transduction by double-stranded adeno-associated virus vectors in vitro and in vivo. Gene Ther. 2003;10:2105–2111. doi: 10.1038/sj.gt.3302133. [DOI] [PubMed] [Google Scholar]
  • 16.Everett RS, Evans HK, Hodges BL, Ding EY, Serra DM, Amalfitano A. Strain-specific rate of shutdown of cmv enhancer activity in murine liver confirmed by use of persistent [e1(−), e2b(−)] adenoviral vectors. Virology. 2004;325:96–105. doi: 10.1016/j.virol.2004.04.032. [DOI] [PubMed] [Google Scholar]
  • 17.Brooks AR, Harkins RN, Wang P, Qian HS, Liu P, Rubanyi GM. Transcriptional silencing is associated with extensive methylation of the cmv promoter following adenoviral gene delivery to muscle. J Gene Med. 2004;6:395–404. doi: 10.1002/jgm.516. [DOI] [PubMed] [Google Scholar]
  • 18.Subramaniam A, Jones WK, Gulick J, Wert S, Neumann J, Robbins J. Tissue-specific regulation of the alpha-myosin heavy chain gene promoter in transgenic mice. J Biol Chem. 1991;266:24613–24620. [PubMed] [Google Scholar]
  • 19.Griscelli F, Gilardi-Hebenstreit P, Hanania N, Franz WM, Opolon P, Perricaudet M, Ragot T. Heart-specific targeting of beta-galactosidase by the ventricle-specific cardiac myosin light chain 2 promoter using adenovirus vectors. Hum Gene Ther. 1998;9:1919–1928. doi: 10.1089/hum.1998.9.13-1919. [DOI] [PubMed] [Google Scholar]
  • 20.Prasad KM, Xu Y, Yang Z, Acton ST, French BA. Robust cardiomyocyte-specific gene expression following systemic injection of aav: In vivo gene delivery follows a poisson distribution. Gene Ther. 2011;18:43–52. doi: 10.1038/gt.2010.105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Fleury S, Simeoni E, Zuppinger C, Deglon N, von Segesser LK, Kappenberger L, Vassalli G. Multiply attenuated, self-inactivating lentiviral vectors efficiently deliver and express genes for extended periods of time in adult rat cardiomyocytes in vivo. Circulation. 2003;107:2375–2382. doi: 10.1161/01.CIR.0000065598.46411.EF. [DOI] [PubMed] [Google Scholar]
  • 22.Tilemann L, Ishikawa K, Weber T, Hajjar RJ. Gene therapy for heart failure. Circ Res. 2012;110:777–793. doi: 10.1161/CIRCRESAHA.111.252981. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Katz MG, Swain JD, Tomasulo CE, Sumaroka M, Fargnoli A, Bridges CR. Current strategies for myocardial gene delivery. J Mol Cell Cardiol. 2011;50:766–776. doi: 10.1016/j.yjmcc.2010.09.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Lai NC, Roth DM, Gao MH, Fine S, Head BP, Zhu J, McKirnan MD, Kwong C, Dalton N, Urasawa K, Roth DA, Hammond HK. Intracoronary delivery of adenovirus encoding adenylyl cyclase vi increases left ventricular function and camp-generating capacity. Circulation. 2000;102:2396–2401. doi: 10.1161/01.cir.102.19.2396. [DOI] [PubMed] [Google Scholar]
  • 25.Kaspar BK, Roth DM, Lai NC, Drumm JD, Erickson DA, McKirnan MD, Hammond HK. Myocardial gene transfer and long-term expression following intracoronary delivery of adeno-associated virus. J Gene Med. 2005;7:316–324. doi: 10.1002/jgm.665. [DOI] [PubMed] [Google Scholar]
  • 26.Roth DM, Lai NC, Gao MH, Drumm JD, Jimenez J, Feramisco JR, Hammond HK. Indirect intracoronary delivery of adenovirus encoding adenylyl cyclase increases left ventricular contractile function in mice. Am J Physiol Heart Circ Physiol. 2004;287:H172–H177. doi: 10.1152/ajpheart.01009.2003. [DOI] [PubMed] [Google Scholar]
  • 27.Lai NC, Roth DM, Gao MH, Tang T, Dalton N, Lai YY, Spellman M, Clopton P, Hammond HK. Intracoronary adenovirus encoding adenylyl cyclase vi increases left ventricular function in heart failure. Circulation. 2004;110:330–336. doi: 10.1161/01.CIR.0000136033.21777.4D. [DOI] [PubMed] [Google Scholar]
  • 28.Roth DM, Lai NC, Gao MH, Fine S, McKirnan MD, Roth DA, Hammond HK. Nitroprusside increases gene transfer associated with intracoronary delivery of adenovirus. Hum Gene Ther. 2004;15:989–994. doi: 10.1089/hum.2004.15.989. [DOI] [PubMed] [Google Scholar]
  • 29.Donahue JK, Heldman AW, Fraser H, McDonald AD, Miller JM, Rade JJ, Eschenhagen T, Marban E. Focal modification of electrical conduction in the heart by viral gene transfer. Nat Med. 2000;6:1395–1398. doi: 10.1038/82214. [DOI] [PubMed] [Google Scholar]
  • 30.Iwatate M, Gu Y, Dieterle T, Iwanaga Y, Peterson KL, Hoshijima M, Chien KR, Ross J. In vivo high-efficiency transcoronary gene delivery and cre-loxp gene switching in the adult mouse heart. Gene Ther. 2003;10:1814–1820. doi: 10.1038/sj.gt.3302077. [DOI] [PubMed] [Google Scholar]
  • 31.Davis J, Westfall MV, Townsend D, Blankinship M, Herron TJ, Guerrero-Serna G, Wang W, Devaney E, Metzger JM. Designing heart performance by gene transfer. Physiol Rev. 2008;88:1567–1651. doi: 10.1152/physrev.00039.2007. [DOI] [PubMed] [Google Scholar]
  • 32.Inagaki K, Fuess S, Storm TA, Gibson GA, McTiernan CF, Kay MA, Nakai H. Robust systemic transduction with aav9 vectors in mice: Efficient global cardiac gene transfer superior to that of aav8. Mol Ther. 2006;14:45–53. doi: 10.1016/j.ymthe.2006.03.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Fang H, Lai NC, Gao MH, Roth DM, Miyanohara A, Tang T, Hammond HK. Intracoronary vs intravenous delivery of adeno-associated virus for cardiac gene transfer in mice. Hum Gene Ther Mothods. 2012 doi: 10.1089/hgtb.2012.105. in press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Hoppe UC, Marban E, Johns DC. Adenovirus-mediated inducible gene expression in vivo by a hybrid ecdysone receptor. Mol Ther. 2000;1:159–164. doi: 10.1006/mthe.1999.0023. [DOI] [PubMed] [Google Scholar]
  • 35.Sipo I, Wang X, Hurtado Pico A, Suckau L, Weger S, Poller W, Fechner H. Tamoxifen-regulated adenoviral e1a chimeras for the control of tumor selective oncolytic adenovirus replication in vitro and in vivo. Gene Ther. 2006;13:173–186. doi: 10.1038/sj.gt.3302604. [DOI] [PubMed] [Google Scholar]
  • 36.Goverdhana S, Puntel M, Xiong W, Zirger JM, Barcia C, Curtin JF, Soffer EB, Mondkar S, King GD, Hu J, Sciascia SA, Candolfi M, Greengold DS, Lowenstein PR, Castro MG. Regulatable gene expression systems for gene therapy applications: Progress and future challenges. Mol Ther. 2005;12:189–211. doi: 10.1016/j.ymthe.2005.03.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Rivera VM, Clackson T, Natesan S, Pollock R, Amara JF, Keenan T, Magari SR, Phillips T, Courage NL, Cerasoli F, Jr, Holt DA, Gilman M. A humanized system for pharmacologic control of gene expression. Nat Med. 1996;2:1028–1032. doi: 10.1038/nm0996-1028. [DOI] [PubMed] [Google Scholar]
  • 38.Stieger K, Le Meur G, Lasne F, Weber M, Deschamps JY, Nivard D, Mendes-Madeira A, Provost N, Martin L, Moullier P, Rolling F. Long-term doxycycline-regulated transgene expression in the retina of nonhuman primates following subretinal injection of recombinant aav vectors. Mol Ther. 2006;13:967–975. doi: 10.1016/j.ymthe.2005.12.001. [DOI] [PubMed] [Google Scholar]
  • 39.Stieger K, Belbellaa B, Le Guiner C, Moullier P, Rolling F. In vivo gene regulation using tetracycline-regulatable systems. Adv Drug Deliv Rev. 2009;61:527–541. doi: 10.1016/j.addr.2008.12.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Harrison DE, Strong R, Sharp ZD, Nelson JF, Astle CM, Flurkey K, Nadon NL, Wilkinson JE, Frenkel K, Carter CS, Pahor M, Javors MA, Fernandez E, Miller RA. Rapamycin fed late in life extends lifespan in genetically heterogeneous mice. Nature. 2009;460:392–395. doi: 10.1038/nature08221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Villarreal FJ, Griffin M, Omens J, Dillmann W, Nguyen J, Covell J. Early short-term treatment with doxycycline modulates postinfarction left ventricular remodeling. Circulation. 2003;108:1487–1492. doi: 10.1161/01.CIR.0000089090.05757.34. [DOI] [PubMed] [Google Scholar]
  • 42.Stieger K, Mendes-Madeira A, Meur GL, Weber M, Deschamps JY, Nivard D, Provost N, Moullier P, Rolling F. Oral administration of doxycycline allows tight control of transgene expression: A key step towards gene therapy of retinal diseases. Gene Ther. 2007;14:1668–1673. doi: 10.1038/sj.gt.3303034. [DOI] [PubMed] [Google Scholar]
  • 43.Gao MH, Bayat H, Roth DM, Yao Zhou J, Drumm J, Burhan J, Hammond HK. Controlled expression of cardiac-directed adenylylcyclase type vi provides increased contractile function. Cardiovasc Res. 2002;56:197–204. doi: 10.1016/s0008-6363(02)00539-4. [DOI] [PubMed] [Google Scholar]
  • 44.Rolain JM, Mallet MN, Raoult D. Correlation between serum doxycycline concentrations and serologic evolution in patients with coxiella burnetii endocarditis. J Infect Dis. 2003;188:1322–1325. doi: 10.1086/379082. [DOI] [PubMed] [Google Scholar]
  • 45.Berman B, Perez OA, Zell D. Update on rosacea and anti-inflammatory-dose doxycycline. Drugs Today (Barc) 2007;43:27–34. doi: 10.1358/dot.2007.43.1.1025697. [DOI] [PubMed] [Google Scholar]
  • 46.Lai NC, Tang T, Gao MH, Saito M, Miyanohara A, Hammond HK. Improved function of the failing rat heart by regulated expression of insulin-like growth factor i via intramuscular gene transfer. Hum Gene Ther. 2012;23:255–261. doi: 10.1089/hum.2011.094. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Herzog RW, Hagstrom JN, Kung SH, Tai SJ, Wilson JM, Fisher KJ, High KA. Stable gene transfer and expression of human blood coagulation factor ix after intramuscular injection of recombinant adeno-associated virus. Proc Natl Acad Sci U S A. 1997;94:5804–5809. doi: 10.1073/pnas.94.11.5804. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Minniti G, Muni R, Lanzetta G, Marchetti P, Enrici RM. Chemotherapy for glioblastoma: Current treatment and future perspectives for cytotoxic and targeted agents. Anticancer Res. 2009;29:5171–5184. [PubMed] [Google Scholar]
  • 49.Zoncu R, Efeyan A, Sabatini DM. Mtor: From growth signal integration to cancer, diabetes and ageing. Nat Rev Mol Cell Biol. 2011;12:21–35. doi: 10.1038/nrm3025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Yang W, Digits CA, Hatada M, Narula S, Rozamus LW, Huestis CM, Wong J, Dalgarno D, Holt DA. Selective epimerization of rapamycin via a retroaldol/aldol mechanism mediated by titanium tetraisopropoxide. Org Lett. 1999;1:2033–2035. doi: 10.1021/ol991209o. [DOI] [PubMed] [Google Scholar]
  • 51.Abraham RT, Wiederrecht GJ. Immunopharmacology of rapamycin. Annu Rev Immunol. 1996;14:483–510. doi: 10.1146/annurev.immunol.14.1.483. [DOI] [PubMed] [Google Scholar]
  • 52.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. doi: 10.1056/NEJM198207223070401. [DOI] [PubMed] [Google Scholar]
  • 53.Rockman HA, Koch WJ, Lefkowitz RJ. Seven-transmembrane-spanning receptors and heart function. Nature. 2002;415:206–212. doi: 10.1038/415206a. [DOI] [PubMed] [Google Scholar]
  • 54.Bristow MR. Treatment of chronic heart failure with beta-adrenergic receptor antagonists: A convergence of receptor pharmacology and clinical cardiology. Circ Res. 2011;109:1176–1194. doi: 10.1161/CIRCRESAHA.111.245092. [DOI] [PubMed] [Google Scholar]
  • 55.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. doi: 10.1161/01.cir.87.2.454. [DOI] [PubMed] [Google Scholar]
  • 56.Raake PW, Vinge LE, Gao E, Boucher M, Rengo G, Chen X, DeGeorge BR, Jr, Matkovich S, Houser SR, Most P, Eckhart AD, Dorn GW, 2nd, Koch WJ. G protein-coupled receptor kinase 2 ablation in cardiac myocytes before or after myocardial infarction prevents heart failure. Circ Res. 2008;103:413–422. doi: 10.1161/CIRCRESAHA.107.168336. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.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 U S A. 2000;97:5428–5433. doi: 10.1073/pnas.090091197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Rengo G, Lymperopoulos A, Zincarelli C, Donniacuo M, Soltys S, Rabinowitz JE, Koch WJ. Myocardial adeno-associated virus serotype 6-betaarkct gene therapy improves cardiac function and normalizes the neurohormonal axis in chronic heart failure. Circulation. 2009;119:89–98. doi: 10.1161/CIRCULATIONAHA.108.803999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Williams ML, Hata JA, Schroder J, Rampersaud E, Petrofski J, Jakoi A, Milano CA, Koch WJ. Targeted beta-adrenergic receptor kinase (betaark1) inhibition by gene transfer in failing human hearts. Circulation. 2004;109:1590–1593. doi: 10.1161/01.CIR.0000125521.40985.28. [DOI] [PubMed] [Google Scholar]
  • 60.Harding VB, Jones LR, Lefkowitz RJ, Koch WJ, Rockman HA. Cardiac beta ark1 inhibition prolongs survival and augments beta blocker therapy in a mouse model of severe heart failure. Proc Natl Acad Sci U S A. 2001;98:5809–5814. doi: 10.1073/pnas.091102398. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Sunahara RK, Dessauer CW, Gilman AG. Complexity and diversity of mammalian adenylyl cyclases. Annu Rev Pharmacol Toxicol. 1996;36:461–480. doi: 10.1146/annurev.pa.36.040196.002333. [DOI] [PubMed] [Google Scholar]
  • 62.Pierce KL, Premont RT, Lefkowitz RJ. Seven-transmemrane receptors. Nat Rev Mol Cell Biol. 2002;3:639–650. doi: 10.1038/nrm908. [DOI] [PubMed] [Google Scholar]
  • 63.Marian AJ. Beta-adrenergic receptors signaling and heart failure in mice, rabbits and humans. J Mol Cell Cardiol. 2006;41:11–13. doi: 10.1016/j.yjmcc.2006.04.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Gao MH, Lai NC, Roth DM, Zhou J, Zhu J, Anzai T, Dalton N, Hammond HK. Adenylylcyclase increases responsiveness to catecholamine stimulation in transgenic mice. Circulation. 1999;99:1618–1622. doi: 10.1161/01.cir.99.12.1618. [DOI] [PubMed] [Google Scholar]
  • 65.Feldman AM. Adenylyl cyclase: A new target for heart failure therapeutics. Circulation. 2002;105:1876–1878. doi: 10.1161/01.cir.0000016965.24080.12. [DOI] [PubMed] [Google Scholar]
  • 66.Ping P, Anzai T, Gao M, Hammond HK. Adenylyl cyclase and g protein receptor kinase expression during development of heart failure. Am J Physiol Heart Circ Physiol. 1997;273:H707–H717. doi: 10.1152/ajpheart.1997.273.2.H707. [DOI] [PubMed] [Google Scholar]
  • 67.Ishikawa Y, Sorota S, Kiuchi K, Shannon RP, Komamura K, Katsushika S, Vatner DE, Vatner SF, Homcy CJ. Downregulation of adenylylcyclase types v and vi mrna levels in pacing-induced heart failure in dogs. J Clin Invest. 1994;93:2224–2229. doi: 10.1172/JCI117219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Feldman MD, Copelas L, Gwathmey JK, Phillips P, Warren SE, Schoen FJ, Grossman W, Morgan JP. Deficient production of cyclic amp: Pharmacologic evidence of an important cause of contractile dysfunction in patients with end-stage heart failure. Circulation. 1987;75:331–339. doi: 10.1161/01.cir.75.2.331. [DOI] [PubMed] [Google Scholar]
  • 69.Roth DM, Bayat H, Drumm JD, Gao MH, Swaney JS, Ander A, Hammond HK. Adenylyl cyclase increases survival in cardiomyopathy. Circulation. 2002;105:1989–1994. doi: 10.1161/01.cir.0000014968.54967.d3. [DOI] [PubMed] [Google Scholar]
  • 70.Roth DM, Gao MH, Lai NC, Drumm J, Dalton N, Zhou JY, Zhu J, Entrikin D, Hammond HK. Cardiac-directed adenylyl cyclase expression improves heart function in murine cardiomyopathy. Circulation. 1999;99:3099–3102. doi: 10.1161/01.cir.99.24.3099. [DOI] [PubMed] [Google Scholar]
  • 71.Tang T, Gao MH, Roth DM, Guo T, Hammond HK. Adenylyl cyclase type vi corrects cardiac sarcoplasmic reticulum calcium uptake defects in cardiomyopathy. Am J Physiol Heart Circ Physiol. 2004;287:H1906–H1912. doi: 10.1152/ajpheart.00356.2004. [DOI] [PubMed] [Google Scholar]
  • 72.Roth DM, Drumm JD, Bhargava V, Swaney JS, Gao MH, Hammond HK. Cardiac-directed expression of adenylyl cyclase and heart rate regulation. Basic Res Cardiol. 2003;98:380–387. doi: 10.1007/s00395-003-0429-4. [DOI] [PubMed] [Google Scholar]
  • 73.Tang T, Gao MH, Lai NC, Firth AL, Takahashi T, Guo T, Yuan JX, Roth DM, Hammond HK. Adenylyl cyclase type 6 deletion decreases left ventricular function via impaired calcium handling. Circulation. 2008;117:61–69. doi: 10.1161/CIRCULATIONAHA.107.730069. [DOI] [PubMed] [Google Scholar]
  • 74.Timofeyev V, He Y, Tuteja D, Zhang Q, Roth DM, Hammond HK, Chiamvimonvat N. Cardiac-directed expression of adenylyl cyclase reverses electrical remodeling in cardiomyopathy. J Mol Cell Cardiol. 2006;41:170–181. doi: 10.1016/j.yjmcc.2006.04.008. [DOI] [PubMed] [Google Scholar]
  • 75.Takahashi T, Tang T, Lai NC, Roth DM, Rebolledo B, Saito M, Lew WY, Clopton P, Hammond HK. Increased cardiac adenylyl cyclase expression is associated with increased survival after myocardial infarction. Circulation. 2006;114:388–396. doi: 10.1161/CIRCULATIONAHA.106.632513. [DOI] [PubMed] [Google Scholar]
  • 76.Tang T, Hammond HK, Firth A, Yang Y, Gao MH, Yuan JX, Lai NC. Adenylyl cyclase 6 improves calcium uptake and left ventricular function in aged hearts. J Am Coll Cardiol. 2011;57:1846–1855. doi: 10.1016/j.jacc.2010.11.052. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Bers DM. Altered cardiac myocyte ca regulation in heart failure. Physiology (Bethesda) 2006;21:380–387. doi: 10.1152/physiol.00019.2006. [DOI] [PubMed] [Google Scholar]
  • 78.Gwathmey JK, Copelas L, MacKinnon R, Schoen FJ, Feldman MD, Grossman W, Morgan JP. Abnormal intracellular calcium handling in myocardium from patients with end-stage heart failure. Circ Res. 1987;61:70–76. doi: 10.1161/01.res.61.1.70. [DOI] [PubMed] [Google Scholar]
  • 79.Hasenfuss G, Reinecke H, Studer R, Meyer M, Pieske B, Holtz J, Holubarsch C, Posival H, Just H, Drexler H. Relation between myocardial function and expression of sarcoplasmic reticulum ca(2+)-atpase in failing and nonfailing human myocardium. Circ Res. 1994;75:434–442. doi: 10.1161/01.res.75.3.434. [DOI] [PubMed] [Google Scholar]
  • 80.He H, Giordano FJ, Hilal-Dandan R, Choi DJ, Rockman HA, McDonough PM, Bluhm WF, Meyer M, Sayen MR, Swanson E, Dillmann WH. Overexpression of the rat sarcoplasmic reticulum ca2+ atpase gene in the heart of transgenic mice accelerates calcium transients and cardiac relaxation. J Clin Invest. 1997;100:380–389. doi: 10.1172/JCI119544. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Baker DL, Hashimoto K, Grupp IL, Ji Y, Reed T, Loukianov E, Grupp G, Bhagwhat A, Hoit B, Walsh R, Marban E, Periasamy M. Targeted overexpression of the sarcoplasmic reticulum ca2+-atpase increases cardiac contractility in transgenic mouse hearts. Circ Res. 1998;83:1205–1214. doi: 10.1161/01.res.83.12.1205. [DOI] [PubMed] [Google Scholar]
  • 82.Periasamy M, Reed TD, Liu LH, Ji Y, Loukianov E, Paul RJ, Nieman ML, Riddle T, Duffy JJ, Doetschman T, Lorenz JN, Shull GE. Impaired cardiac performance in heterozygous mice with a null mutation in the sarco(endo)plasmic reticulum ca2+-atpase isoform 2 (serca2) gene. J Biol Chem. 1999;274:2556–2562. doi: 10.1074/jbc.274.4.2556. [DOI] [PubMed] [Google Scholar]
  • 83.Schultz Jel J, Glascock BJ, Witt SA, Nieman ML, Nattamai KJ, Liu LH, Lorenz JN, Shull GE, Kimball TR, Periasamy M. Accelerated onset of heart failure in mice during pressure overload with chronically decreased serca2 calcium pump activity. Am J Physiol Heart Circ Physiol. 2004;286:H1146–H1153. doi: 10.1152/ajpheart.00720.2003. [DOI] [PubMed] [Google Scholar]
  • 84.del Monte F, Harding SE, Schmidt U, Matsui T, Kang ZB, Dec GW, Gwathmey JK, Rosenzweig A, Hajjar RJ. Restoration of contractile function in isolated cardiomyocytes from failing human hearts by gene transfer of serca2a. Circulation. 1999;100:2308–2311. doi: 10.1161/01.cir.100.23.2308. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Miyamoto MI, del Monte F, Schmidt U, DiSalvo TS, Kang ZB, Matsui T, Guerrero JL, Gwathmey JK, Rosenzweig A, Hajjar RJ. Adenoviral gene transfer of serca2a improves left-ventricular function in aortic-banded rats in transition to heart failure. Proc Natl Acad Sci U S A. 2000;97:793–798. doi: 10.1073/pnas.97.2.793. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.del Monte F, Williams E, Lebeche D, Schmidt U, Rosenzweig A, Gwathmey JK, Lewandowski ED, Hajjar RJ. Improvement in survival and cardiac metabolism after gene transfer of sarcoplasmic reticulum ca(2+)-atpase in a rat model of heart failure. Circulation. 2001;104:1424–1429. doi: 10.1161/hc3601.095574. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Schmidt U, del Monte F, Miyamoto MI, Matsui T, Gwathmey JK, Rosenzweig A, Hajjar RJ. Restoration of diastolic function in senescent rat hearts through adenoviral gene transfer of sarcoplasmic reticulum ca(2+)-atpase. Circulation. 2000;101:790–796. doi: 10.1161/01.cir.101.7.790. [DOI] [PubMed] [Google Scholar]
  • 88.Byrne MJ, Power JM, Preovolos A, Mariani JA, Hajjar RJ, Kaye DM. Recirculating cardiac delivery of aav2/1serca2a improves myocardial function in an experimental model of heart failure in large animals. Gene Ther. 2008;15:1550–1557. doi: 10.1038/gt.2008.120. [DOI] [PubMed] [Google Scholar]
  • 89.Most P, Bernotat J, Ehlermann P, Pleger ST, Reppel M, Borries M, Niroomand F, Pieske B, Janssen PM, Eschenhagen T, Karczewski P, Smith GL, Koch WJ, Katus HA, Remppis A. S100a1: A regulator of myocardial contractility. Proc Natl Acad Sci U S A. 2001;98:13889–13894. doi: 10.1073/pnas.241393598. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Kiewitz R, Acklin C, Schafer BW, Maco B, Uhrik B, Wuytack F, Erne P, Heizmann CW. Ca2+ -dependent interaction of s100a1 with the sarcoplasmic reticulum ca2+ -atpase2a and phospholamban in the human heart. Biochem Biophys Res Commun. 2003;306:550–557. doi: 10.1016/s0006-291x(03)00987-2. [DOI] [PubMed] [Google Scholar]
  • 91.Treves S, Scutari E, Robert M, Groh S, Ottolia M, Prestipino G, Ronjat M, Zorzato F. Interaction of s100a1 with the ca2+ release channel (ryanodine receptor) of skeletal muscle. Biochemistry. 1997;36:11496–11503. doi: 10.1021/bi970160w. [DOI] [PubMed] [Google Scholar]
  • 92.Remppis A, Greten T, Schafer BW, Hunziker P, Erne P, Katus HA, Heizmann CW. Altered expression of the ca(2+)-binding protein s100a1 in human cardiomyopathy. Biochim Biophys Acta. 1996;1313:253–257. doi: 10.1016/0167-4889(96)00097-3. [DOI] [PubMed] [Google Scholar]
  • 93.Most P, Remppis A, Pleger ST, Loffler E, Ehlermann P, Bernotat J, Kleuss C, Heierhorst J, Ruiz P, Witt H, Karczewski P, Mao L, Rockman HA, Duncan SJ, Katus HA, Koch WJ. Transgenic overexpression of the ca2+-binding protein s100a1 in the heart leads to increased in vivo myocardial contractile performance. J Biol Chem. 2003;278:33809–33817. doi: 10.1074/jbc.M301788200. [DOI] [PubMed] [Google Scholar]
  • 94.Pleger ST, Most P, Boucher M, Soltys S, Chuprun JK, Pleger W, Gao E, Dasgupta A, Rengo G, Remppis A, Katus HA, Eckhart AD, Rabinowitz JE, Koch WJ. Stable myocardial-specific aav6-s100a1 gene therapy results in chronic functional heart failure rescue. Circulation. 2007;115:2506–2515. doi: 10.1161/CIRCULATIONAHA.106.671701. [DOI] [PubMed] [Google Scholar]
  • 95.Brinks H, Rohde D, Voelkers M, Qiu G, Pleger ST, Herzog N, Rabinowitz J, Ruhparwar A, Silvestry S, Lerchenmuller C, Mather PJ, Eckhart AD, Katus HA, Carrel T, Koch WJ, Most P. S100a1 genetically targeted therapy reverses dysfunction of human failing cardiomyocytes. J Am Coll Cardiol. 2011;58:966–973. doi: 10.1016/j.jacc.2011.03.054. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Brinks H, Boucher M, Gao E, Chuprun JK, Pesant S, Raake PW, Huang ZM, Wang X, Qiu G, Gumpert A, Harris DM, Eckhart AD, Most P, Koch WJ. Level of g protein-coupled receptor kinase-2 determines myocardial ischemia/reperfusion injury via pro- and anti-apoptotic mechanisms. Circ Res. 2010;107:1140–1149. doi: 10.1161/CIRCRESAHA.110.221010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Gao MH, Tang T, Guo T, Miyanohara A, Yajima T, Pestonjamasp K, Feramisco JR, Hammond HK. Adenylyl cyclase type vi increases akt activity and phospholamban phosphorylation in cardiac myocytes. J Biol Chem. 2008;283:33527–33535. doi: 10.1074/jbc.M805825200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Most P, Boerries M, Eicher C, Schweda C, Ehlermann P, Pleger ST, Loeffler E, Koch WJ, Katus HA, Schoenenberger CA, Remppis A. Extracellular s100a1 protein inhibits apoptosis in ventricular cardiomyocytes via activation of the extracellular signal-regulated protein kinase 1/2 (erk1/2) J Biol Chem. 2003;278:48404–48412. doi: 10.1074/jbc.M308587200. [DOI] [PubMed] [Google Scholar]
  • 99.Hulot JS, Senyei G, Hajjar RJ. Sarcoplasmic reticulum and calcium cycling targeting by gene therapy. Gene Ther. 2012;19:596–599. doi: 10.1038/gt.2012.34. [DOI] [PubMed] [Google Scholar]
  • 100.Rohde D, Ritterhoff J, Voelkers M, Katus HA, Parker TG, Most P. S100a1: A multifaceted therapeutic target in cardiovascular disease. J Cardiovasc Transl Res. 2010;3:525–537. doi: 10.1007/s12265-010-9211-9. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES