Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Lisa Tilemann is active.

Publication


Featured researches published by Lisa Tilemann.


Circulation Research | 2012

Gene Therapy for Heart Failure

Lisa Tilemann; Kiyotake Ishikawa; Thomas Weber; Roger J. Hajjar

Congestive heart failure accounts for half a million deaths per year in the United States. Despite its place among the leading causes of morbidity, pharmacological and mechanic remedies have only been able to slow the progression of the disease. Todays science has yet to provide a cure, and there are few therapeutic modalities available for patients with advanced heart failure. There is a critical need to explore new therapeutic approaches in heart failure, and gene therapy has emerged as a viable alternative. Recent advances in understanding of the molecular basis of myocardial dysfunction, together with the evolution of increasingly efficient gene transfer technology, have placed heart failure within reach of gene-based therapy. The recent successful and safe completion of a phase 2 trial targeting the sarcoplasmic reticulum calcium ATPase pump (SERCA2a), along with the start of more recent phase 1 trials, opens a new era for gene therapy for the treatment of heart failure.


Circulation-heart Failure | 2013

AAV9.I-1c Delivered via Direct Coronary Infusion in a Porcine Model of Heart Failure Improves Contractility and Mitigates Adverse Remodeling

Kenneth Fish; Dennis Ladage; Yoshiaki Kawase; Ioannis Karakikes; Dongtak Jeong; Hung Ly; Kiyotake Ishikawa; Lahouaria Hadri; Lisa Tilemann; Jochen Müller-Ehmsen; R. Jude Samulski; Evangelia G. Kranias; Roger J. Hajjar

Background—Heart failure is characterized by impaired function and disturbed Ca2+ homeostasis. Transgenic increases in inhibitor-1 activity have been shown to improve Ca2 cycling and preserve cardiac performance in the failing heart. The aim of this study was to evaluate the effect of activating the inhibitor (I-1c) of protein phosphatase 1 (I-1) through gene transfer on cardiac function in a porcine model of heart failure induced by myocardial infarction. Methods and Results—Myocardial infarction was created by a percutaneous, permanent left anterior descending artery occlusion in Yorkshire Landrace swine (n=16). One month after myocardial infarction, pigs underwent intracoronary delivery of either recombinant adeno-associated virus type 9 carrying I-1c (n=8) or saline (n=6) as control. One month after myocardial infarction was created, animals exhibited severe heart failure demonstrated by decreased ejection fraction (46.4±7.0% versus sham 69.7±8.5%) and impaired (dP/dt)max and (dP/dt)min. Intracoronary injection of AAV9.I-1c prevented further deterioration of cardiac function and led to a decrease in scar size. Conclusions—In this preclinical model of heart failure, overexpression of I-1c by intracoronary in vivo gene transfer preserved cardiac function and reduced the scar size.


Science Translational Medicine | 2013

SUMO-1 Gene Transfer Improves Cardiac Function in a Large-Animal Model of Heart Failure

Lisa Tilemann; Ahyoung Lee; Kiyotake Ishikawa; Jaume Aguero; Kleopatra Rapti; Carlos G. Santos-Gallego; Erik Kohlbrenner; Kenneth Fish; Changwon Kho; Roger J. Hajjar

Cardiac gene delivery of small ubiquitin-related modifier 1 (SUMO-1) improved cardiac function and stabilized left ventricular volumes in a swine model of ischemic heart failure. Cardiac Gene Therapy to the Rescue Heart failure (HF) is one of the top reasons for hospitalization among the elderly and remains a leading cause of death in the Western world. Gene therapy has been proposed as a way to coerce the heart into being healthy by targeting cardiac-specific pathways. Replacing the gene sarcoplasmic reticulum Ca2+ adenosine triphosphatase (SERCA2a) in patients has made it to phase 2b/3 trials, with early signs pointing to an improvement in HF-related events. To boost the effects of SERCA2a, Tilemann et al. designed a large-animal study that also tests the delivery of small ubiquitin-related modifier 1 (SUMO-1)—an important regulator of SERCA2a. The authors compared the efficacy of SUMO-1 gene transfer to SERCA2a gene transfer alone and to the combined delivery of both genes in a pig model of HF. In addition to being safe, administering SUMO-1 directly to the heart of these animals showed improved cardiac contractility and prevented left ventricular dilatation (two major aspects of HF). According to the authors, the functional improvements in this model of heart failure are most likely the result of improved SR Ca2+ ATPase activity afforded by increased SUMO-1 protein levels. Delivery of both SUMO-1 and SERCA2a suggested additional beneficial effects, but more mechanistic studies will be needed to understand this potential synergy. With the precedent set by the SERCA2a clinical trials, moving SUMO-1 gene therapy from pigs to humans seems likely in the short-term. Recently, the impact of small ubiquitin-related modifier 1 (SUMO-1) on the regulation and preservation of sarcoplasmic reticulum calcium adenosine triphosphatase (SERCA2a) function was discovered. The amount of myocardial SUMO-1 is decreased in failing hearts, and its knockdown results in severe heart failure (HF) in mice. In a previous study, we showed that SUMO-1 gene transfer substantially improved cardiac function in a murine model of pressure overload–induced HF. Toward clinical translation, we evaluated in this study the effects of SUMO-1 gene transfer in a swine model of ischemic HF. One month after balloon occlusion of the proximal left anterior descending artery followed by reperfusion, the animals were randomized to receive either SUMO-1 at two doses, SERCA2a, or both by adeno-associated vector type 1 (AAV1) gene transfer via antegrade coronary infusion. Control animals received saline infusions. After gene delivery, there was a significant increase in the maximum rate of pressure rise [dP/dt(max)] that was most pronounced in the group that received both SUMO-1 and SERCA2a. The left ventricular ejection fraction (LVEF) improved after high-dose SUMO-1 with or without SERCA2a gene delivery, whereas there was a decline in LVEF in the animals receiving saline. Furthermore, the dilatation of LV volumes was prevented in the treatment groups. SUMO-1 gene transfer therefore improved cardiac function and stabilized LV volumes in a large-animal model of HF. These results support the critical role of SUMO-1 in SERCA2a function and underline the therapeutic potential of SUMO-1 for HF patients.


Molecular Therapy | 2014

Cardiac I-1c Overexpression With Reengineered AAV Improves Cardiac Function in Swine Ischemic Heart Failure

Kiyotake Ishikawa; Kenneth Fish; Lisa Tilemann; Kleopatra Rapti; Jaume Aguero; Carlos G. Santos-Gallego; Ahyoung Lee; Ioannis Karakikes; Chaoqin Xie; Fadi G. Akar; Yuichi J. Shimada; Judith K. Gwathmey; Aravind Asokan; Scott W.J. McPhee; Jade Samulski; Richard Jude Samulski; Daniel C. Sigg; Thomas Weber; Evangelia G. Kranias; Roger J. Hajjar

Cardiac gene therapy has emerged as a promising option to treat advanced heart failure (HF). Advances in molecular biology and gene targeting approaches are offering further novel options for genetic manipulation of the cardiovascular system. The aim of this study was to improve cardiac function in chronic HF by overexpressing constitutively active inhibitor-1 (I-1c) using a novel cardiotropic vector generated by capsid reengineering of adeno-associated virus (BNP116). One month after a large anterior myocardial infarction, 20 Yorkshire pigs randomly received intracoronary injection of either high-dose BNP116.I-1c (1.0 × 10(13) vector genomes (vg), n = 7), low-dose BNP116.I-1c (3.0 × 10(12) vg, n = 7), or saline (n = 6). Compared to baseline, mean left ventricular ejection fraction increased by 5.7% in the high-dose group, and by 5.2% in the low-dose group, whereas it decreased by 7% in the saline group. Additionally, preload-recruitable stroke work obtained from pressure-volume analysis demonstrated significantly higher cardiac performance in the high-dose group. Likewise, other hemodynamic parameters, including stroke volume and contractility index indicated improved cardiac function after the I-1c gene transfer. Furthermore, BNP116 showed a favorable gene expression pattern for targeting the heart. In summary, I-1c overexpression using BNP116 improves cardiac function in a clinically relevant model of ischemic HF.


Circulation Research | 2011

Inhibition of PKCα/β With Ruboxistaurin Antagonizes Heart Failure in Pigs After Myocardial Infarction Injury

Dennis Ladage; Lisa Tilemann; Kiyotake Ishikawa; Robert N. Correll; Yoshiaki Kawase; Steven R. Houser; Jeffery D. Molkentin; Roger J. Hajjar

Rationale: Protein kinase C&agr; (PKC&agr;) activity and protein level are induced during cardiac disease where it controls myocardial contractility and propensity to heart failure in mice and rats. For example, mice lacking the gene for PKC&agr; have enhanced cardiac contractility and reduced susceptibility to heart failure after long-term pressure overload or after myocardial infarction injury. Pharmacological inhibition of PKC&agr;/&bgr; with Ro-32-0432, Ro-31-8220 or ruboxistaurin (LY333531) similarly enhances cardiac function and antagonizes heart failure in multiple models of disease in both mice and rats. Objective: Large and small mammals differ in several key indexes of heart function and biochemistry, lending uncertainty as to how PKC&agr;/&bgr; inhibition might affect or protect a large animal model of heart failure. Methods and Results: We demonstrate that ruboxistaurin administration to a pig model of myocardial infarction–induced heart failure was protective. Twenty-kilogram pigs underwent left anterior descending artery occlusion resulting in myocardial infarctions and were then divided into vehicle or ruboxistaurin feed groups, after which they were monitored monthly for the next 3 months. Ruboxistaurin administered pigs showed significantly better recovery of myocardial contractility 3 months after infarction injury, greater ejection fraction, and greater cardiac output compared with vehicle-treated pigs. Conclusions: These results provide additional evidence in a large animal model of disease that PKC&agr;/&bgr; inhibition (with ruboxistaurin) represents a tenable and novel therapeutic approach for treating human heart failure.


PLOS ONE | 2013

Stimulating Myocardial Regeneration with Periostin Peptide in Large Mammals Improves Function Post-Myocardial Infarction but Increases Myocardial Fibrosis

Dennis Ladage; Elisa Yaniz-Galende; Kleopatra Rapti; Kiyotake Ishikawa; Lisa Tilemann; Scott Shapiro; Yoshiaki Takewa; Jochen Müller-Ehmsen; Martin Schwarz; Mario J. Garcia; Javier Sanz; Roger J. Hajjar; Yoshiaki Kawase

Aims Mammalian myocardium has a finite but limited capacity to regenerate. Experimentally stimulating proliferation of cardiomyocytes with extracellular regeneration factors like periostin enhances cardiac repair in rodents. The aim of this study was to develop a safe method for delivering regeneration factors to the heart and to test the functional and structural effects of periostin peptide treatment in a large animal model of myocardial infarction (MI). Methods and Results We developed a controlled release system to deliver recombinant periostin peptide into the pericardial space. A single application of this method was performed two days after experimental MI in swine. Animals were randomly assigned to receive either saline or periostin peptide. Experimental groups were compared at baseline, day 2, 1 month and 3 months. Treatment with periostin peptide increased the EF from 31% to 41% and decreased by 22% the infarct size within 12 weeks. Periostin peptide-treated animals had newly formed myocardium strips within the infarct scar, leading to locally improved myocardial function. In addition the capillary density was increased in animals receiving periostin. However, periostin peptide treatment increased myocardial fibrosis in the remote region at one week and 12 weeks post-treatment. Conclusion Our study shows that myocardial regeneration through targeted peptides is possible. However, in the case of periostin the effects on cardiac fibrosis may limit its clinical application as a viable therapeutic strategy.


Journal of Gene Medicine | 2011

Gene delivery methods in cardiac gene therapy

Kiyotake Ishikawa; Lisa Tilemann; Kenneth Fish; Roger J. Hajjar

Gene therapy for the treatment of heart failure is emerging as a multidisciplinary field demonstrating advances with respect to identifying key signaling pathways, modernized vector creation and delivery technologies. Although these discoveries offer significant progress, selecting optimal methods for the vector delivery remains a key component for efficient cardiac gene therapy to validate the targets in rodent models and to test clinically relevant ones in pre‐clinical models. Although the goals of higher transduction efficiency and cardiac specificity can be achieved with several delivery methods, the invasiveness and patient safety remain unclear for clinical application. In this review, we discuss various features of the currently available vector delivery methods for cardiac gene therapy. Copyright


Gene Therapy | 2012

Cardiac gene therapy in large animals: bridge from bench to bedside

Kiyotake Ishikawa; Lisa Tilemann; Dennis Ladage; Jaume Aguero; Lauren Leonardson; Kenneth Fish; Yoshiaki Kawase

Several clinical trials are evaluating gene transfer as a therapeutic approach to treat cardiac diseases. Although it has just started on the path to clinical application, recent advances in gene delivery technologies with increasing knowledge of underlying mechanisms raise great expectations for the cardiac gene therapy. Although in vivo experiments using small animals provide the therapeutic potential of gene transfer, there exist many fundamental differences between the small animal and the human hearts. Before applying the therapy to clinical patients, large animal studies are a prerequisite to validate the efficacy in an animal model more relevant to the human heart. Several key factors including vector type, injected dose, delivery method and targeted cardiac disease are all important factors that determine the therapeutic efficacy. Selecting the most optimal combination of these factors is essential for successful gene therapy. In addition to the efficacy, safety profiles need to be addressed as well. In this regard, large animal studies are best suited for comprehensive evaluation at the preclinical stages of therapeutic development to ensure safe and effective gene transfer. As the cardiac gene therapy expands its potential, large animal studies will become more important to bridge the bench side knowledge to the clinical arena.


American Journal of Physiology-heart and Circulatory Physiology | 2012

Assessing left ventricular systolic dysfunction after myocardial infarction: are ejection fraction and dP/dtmax complementary or redundant?

Kiyotake Ishikawa; Elie R. Chemaly; Lisa Tilemann; Kenneth Fish; Dennis Ladage; Jaime Aguero; Torsten Vahl; Carlos G. Santos-Gallego; Yoshiaki Kawase; Roger J. Hajjar

Among the various cardiac contractility parameters, left ventricular (LV) ejection fraction (EF) and maximum dP/dt (dP/dt(max)) are the simplest and most used. However, these parameters are often reported together, and it is not clear if they are complementary or redundant. We sought to compare the discriminative value of EF and dP/dt(max) in assessing systolic dysfunction after myocardial infarction (MI) in swine. A total of 220 measurements were obtained. All measurements included LV volumes and EF analysis by left ventriculography, invasive ventricular pressure tracings, and echocardiography. Baseline measurements were performed in 132 pigs, and 88 measurements were obtained at different time points after MI creation. Receiver operator characteristic (ROC) curves to distinguish the presence or absence of an MI revealed a good predictive value for EF [area under the curve (AUC): 0.998] but not by dP/dt(max) (AUC: 0.69, P < 0.001 vs. EF). Dividing dP/dt(max) by LV end-diastolic pressure and heart rate (HR) significantly increased the AUC to 0.87 (P < 0.001 vs. dP/dt(max) and P < 0.001 vs. EF). In naïve pigs, the coefficient of variation of dP/dt(max) was twice than that of EF (22.5% vs. 9.5%, respectively). Furthermore, in n = 19 pigs, dP/dt(max) increased after MI. However, echocardiographic strain analysis of 23 pigs with EF ranging only from 36% to 40% after MI revealed significant correlations between dP/dt(max) and strain parameters in the noninfarcted area (circumferential strain: r = 0.42, P = 0.05; radial strain: r = 0.71, P < 0.001). In conclusion, EF is a more accurate measure of systolic dysfunction than dP/dt(max) in a swine model of MI. Despite the variability of dP/dt(max) both in naïve pigs and after MI, it may sensitively reflect the small changes of myocardial contractility.


Circulation-heart Failure | 2015

Stem Cell Factor Gene Transfer Improves Cardiac Function After Myocardial Infarction in Swine

Kiyotake Ishikawa; Kenneth Fish; Jaume Aguero; Elisa Yaniz-Galende; Dongtak Jeong; Changwon Kho; Lisa Tilemann; Lauren Fish; Lifan Liang; Ahmed A. Eltoukhy; Daniel G. Anderson; Krisztina Zsebo; Kevin D. Costa; Roger J. Hajjar

Background—Stem cell factor (SCF), a ligand of the c-kit receptor, is a critical cytokine, which contributes to cell migration, proliferation, and survival. It has been shown that SCF expression increases after myocardial infarction (MI) and may be involved in cardiac repair. The aim of this study was to determine whether gene transfer of membrane-bound human SCF improves cardiac function in a large animal model of MI. Methods and Results—A transmural MI was created by implanting an embolic coil in the left anterior descending artery in Yorkshire pigs. One week after the MI, the pigs received direct intramyocardial injections of either a recombinant adenovirus encoding for SCF (Ad.SCF, n=9) or &bgr;-gal (Ad.&bgr;-gal, n=6) into the infarct border area. At 3 months post-MI, ejection fraction increased by 12% relative to baseline after Ad.SCF therapy, whereas it decreased by 4.2% (P=0.004) in pigs treated with Ad.&bgr;-gal. Preload-recruitable stroke work was significantly higher in pigs after SCF treatment (Ad.SCF, 55.5±11.6 mm Hg versus Ad.&bgr;-gal, 31.6±12.6 mm Hg, P=0.005), indicating enhanced cardiac function. Histological analyses confirmed the recruitment of c-kit+ cells as well as a reduced degree of apoptosis 1 week after Ad.SCF injection. In addition, increased capillary density compared with pigs treated with Ad.&bgr;-gal was found at 3 months and suggests an angiogenic role of SCF. Conclusions—Local overexpression of SCF post-MI induces the recruitment of c-kit+ cells at the infarct border area acutely. In the chronic stages, SCF gene transfer was associated with improved cardiac function in a preclinical model of ischemic cardiomyopathy.

Collaboration


Dive into the Lisa Tilemann's collaboration.

Top Co-Authors

Avatar

Kiyotake Ishikawa

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Roger J. Hajjar

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Dennis Ladage

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Yoshiaki Kawase

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Kenneth Fish

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Carlos G. Santos-Gallego

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Kleopatra Rapti

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Dongtak Jeong

Icahn School of Medicine at Mount Sinai

View shared research outputs
Researchain Logo
Decentralizing Knowledge