Network


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

Hotspot


Dive into the research topics where Kiyotake Ishikawa is active.

Publication


Featured researches published by Kiyotake Ishikawa.


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.


Molecular Therapy | 2012

Neutralizing Antibodies Against AAV Serotypes 1, 2, 6, and 9 in Sera of Commonly Used Animal Models

Kleopatra Rapti; Vedell Louis-Jeune; Erik Kohlbrenner; Kiyotake Ishikawa; Dennis Ladage; Sergei Zolotukhin; Roger J. Hajjar; Thomas Weber

Adeno-associated virus (AAV)-based vectors are promising gene delivery vehicles for human gene transfer. One significant obstacle to AAV-based gene therapy is the high prevalence of neutralizing antibodies in humans. Until now, it was thought that, except for nonhuman primates, pre-existing neutralizing antibodies are not a problem in small or large animal models for gene therapy. Here, we demonstrate that sera of several animal models of cardiovascular diseases harbor pre-existing antibodies against the cardiotropic AAV serotypes AAV1, AAV6, and AAV9 and against AAV2. The neutralizing antibody titers vary widely both between species and between serotypes. Of all species tested, rats displayed the lowest levels of neutralizing antibodies. Surprisingly, naive mice obtained directly from commercial vendors harbored neutralizing antibodies. Of the large animal models tested, the neutralization of AAV6 transduction by dog sera was especially pronounced. Sera of sheep and rabbits showed modest neutralization of AAV transduction whereas porcine sera strongly inhibited transduction by all AAV serotypes and displayed the largest variation between individual animals. Importantly, neutralizing antibody titers as low as 1/4 completely prevented in vivo transduction by AAV9 in rats. Our results suggest that prescreening of animals for neutralizing antibodies will be important for future gene transfer experiments in these animal models.


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.


Circulation | 2013

Therapeutic Efficacy of AAV1.SERCA2a in Monocrotaline-Induced Pulmonary Arterial Hypertension

Lahouaria Hadri; Razmig Garo Kratlian; Ludovic Benard; Bradley A. Maron; Peter Dorfmüller; Dennis Ladage; Christophe Guignabert; Kiyotake Ishikawa; Jaume Aguero; Borja Ibanez; Irene C. Turnbull; Erik Kohlbrenner; Lifan Liang; Krisztina Zsebo; Marc Humbert; Jean-Sébastien Hulot; Yoshiaki Kawase; Roger J. Hajjar; Jane A. Leopold

Background— Pulmonary arterial hypertension (PAH) is characterized by dysregulated proliferation of pulmonary artery smooth muscle cells leading to (mal)adaptive vascular remodeling. In the systemic circulation, vascular injury is associated with downregulation of sarcoplasmic reticulum Ca2+-ATPase 2a (SERCA2a) and alterations in Ca2+ homeostasis in vascular smooth muscle cells that stimulate proliferation. We, therefore, hypothesized that downregulation of SERCA2a is permissive for pulmonary vascular remodeling and the development of PAH. Methods and Results— SERCA2a expression was decreased significantly in remodeled pulmonary arteries from patients with PAH and the rat monocrotaline model of PAH in comparison with controls. In human pulmonary artery smooth muscle cells in vitro, SERCA2a overexpression by gene transfer decreased proliferation and migration significantly by inhibiting NFAT/STAT3. Overexpresion of SERCA2a in human pulmonary artery endothelial cells in vitro increased endothelial nitric oxide synthase expression and activation. In monocrotaline rats with established PAH, gene transfer of SERCA2a via intratracheal delivery of aerosolized adeno-associated virus serotype 1 (AAV1) carrying the human SERCA2a gene (AAV1.SERCA2a) decreased pulmonary artery pressure, vascular remodeling, right ventricular hypertrophy, and fibrosis in comparison with monocrotaline-PAH rats treated with a control AAV1 carrying &bgr;-galactosidase or saline. In a prevention protocol, aerosolized AAV1.SERCA2a delivered at the time of monocrotaline administration limited adverse hemodynamic profiles and indices of pulmonary and cardiac remodeling in comparison with rats administered AAV1 carrying &bgr;-galactosidase or saline. Conclusions— Downregulation of SERCA2a plays a critical role in modulating the vascular and right ventricular pathophenotype associated with PAH. Selective pulmonary SERCA2a gene transfer may offer benefit as a therapeutic intervention in PAH.


Molecular Therapy | 2010

SERCA2a Gene Transfer Enhances eNOS Expression and Activity in Endothelial Cells

Lahouaria Hadri; Regis Bobe; Yoshiaki Kawase; Dennis Ladage; Kiyotake Ishikawa; Fabrice Atassi; Djamel Lebeche; Evangelia G. Kranias; Jane A. Leopold; Anne-Marie Lompré; Larissa Lipskaia; Roger J. Hajjar

Congestive heart failure (HF) is associated with impaired endothelium-dependent nitric oxide-mediated vasodilatation. The aim of this study was to examine the effects of sarco/endoplasmic reticulum (ER) Ca(2+)-ATPase 2a (SERCA2a) gene transfer on endothelial function in a swine HF model. Two months after the creation of mitral regurgitation to induce HF, the animals underwent intracoronary injection of adeno-associated virus (AAV) carrying SERCA2a (n = 7) or saline (n = 6). At 4 months, coronary flow (CF) was measured in the mid-portion of the left anterior descending (LAD) artery. In the failing animals, CF was decreased significantly; SERCA2a gene transfer rescued CF to levels observed in sham-group [ml/min/g, 0.47 +/- 0.064 saline versus 0.89 +/- 0.116, SERCA2a; P < 0.05; 1.00 +/- 0. 185 sham P = NS (nonsignificant)]. In coronary arteries from HF animals, SERCA2a and endothelial isoform of nitric oxide synthase (eNOS) protein expression were decreased, but restored to normal levels by SERCA2a gene transfer. In human coronary artery endothelial cells (HCAECs), SERCA2a overexpression increased eNOS expression, phosphorylation, eNOS promoter activity, Ca(2+) storage capacity, and enhanced histamine-induced calcium oscillations, eNOS activity, and cyclic guanosine monophosphate (cGMP) production. Thus, SERCA2a gene transfer increases eNOS expression and activity by modulating calcium homeostasis to improve CF. These findings suggest that SERCA2a gene transfer improves vascular reactivity in the setting of HF.


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.


American Journal of Physiology-heart and Circulatory Physiology | 2014

Characterization of right ventricular remodeling and failure in a chronic pulmonary hypertension model

Jaume Aguero; Kiyotake Ishikawa; Lahouaria Hadri; Carlos G. Santos-Gallego; Kenneth Fish; Nadjib Hammoudi; Antoine H. Chaanine; Samantha Torquato; Charbel Naim; Borja Ibanez; Daniel Pereda; Ana García-Álvarez; Valentin Fuster; Partho P. Sengupta; Jane A. Leopold; Roger J. Hajjar

In pulmonary hypertension (PH), right ventricular (RV) dysfunction and failure is the main determinant of a poor prognosis. We aimed to characterize RV structural and functional differences during adaptive RV remodeling and progression to RV failure in a large animal model of chronic PH. Postcapillary PH was created surgically in swine (n = 21). After an 8- to 14-wk follow-up, two groups were identified based on the development of overt heart failure (HF): PH-NF (nonfailing, n = 12) and PH-HF (n = 8). In both groups, invasive hemodynamics, pressure-volume relationships, and echocardiography confirmed a significant increase in pulmonary pressures and vascular resistance consistent with PH. Histological analysis also demonstrated distal pulmonary arterial (PA) remodeling in both groups. Diastolic dysfunction, defined by a steeper RV end-diastolic pressure-volume relationship and longitudinal strain, was found in the absence of HF as an early marker of RV remodeling. RV contractility was increased in both groups, and RV-PA coupling was preserved in PH-NF animals but impaired in the PH-HF group. RV hypertrophy was present in PH-HF, although there was evidence of increased RV fibrosis in both PH groups. In the PH-HF group, RV sarcoplasmic reticulum Ca(2+)-ATPase2a expression was decreased, and endoplasmic reticulum stress was increased. Aldosterone levels were also elevated in PH-HF. Thus, in the swine pulmonary vein banding model of chronic postcapillary PH, RV remodeling occurs at the structural, histological, and molecular level. Diastolic dysfunction and fibrosis are present in adaptive RV remodeling, whereas the onset of RV failure is associated with RV-PA uncoupling, defective calcium handling, and hyperaldosteronism.


European Heart Journal | 2016

Gene therapy for the treatment of heart failure: promise postponed

Jean-Sébastien Hulot; Kiyotake Ishikawa; Roger J. Hajjar

Gene therapy has emerged as a powerful tool in targeting the molecular mechanisms implicated in heart failure. Refinements in vector technology, including the development of recombinant adeno-associated vectors, have allowed for safe, long-term, and efficient gene transfer to the myocardium. These advancements, coupled with evolving delivery techniques, have placed gene therapy as a viable therapeutic option for patients with heart failure. However, after much promise in early-phase clinical trials, the more recent larger clinical trials have shown disappointing results, thus forcing the field to re-evaluate current vectors, delivery systems, targets, and endpoints. We provide here an updated review of current cardiac gene therapy programmes that have been or are being translated into clinical trials.


Gene Therapy | 2011

Delivery of gelfoam-enabled cells and vectors into the pericardial space using a percutaneous approach in a porcine model

Dennis Ladage; Irene C. Turnbull; Kiyotake Ishikawa; Yoshiaki Takewa; Kleopatra Rapti; Charlotte Morel; Ioannis Karakikes; Lahouaria Hadri; Jochen Müller-Ehmsen; Kevin D. Costa; Roger J. Hajjar; Yoshiaki Kawase

Intrapericardial drug delivery is a promising procedure, with the ability to localize therapeutics with the heart. Gelfoam particles are nontoxic, inexpensive, nonimmunogenic and biodegradable compounds that can be used to deliver therapeutic agents. We developed a new percutaneous approach method for intrapericardial injection, puncturing the pericardial sac safely under fluoroscopy and intravascular ultrasound (IVUS) guidance. In a porcine model of myocardial infarction (MI), we deployed gelfoam particles carrying either (a) autologous mesenchymal stem cells (MSCs) or (b) an adenovirus encoding enhanced green fluorescent protein (eGFP) 48 h post-MI. The presence of MSCs and viral infection at the infarct zone was confirmed by immunoflourescence and PCR. Puncture was performed successfully in 16 animals. Using IVUS, we successfully determined the size of the pericardial space before the puncture, and safely accessed that space in setting of pericardial effusion and also adhesions induced by the MI. Intrapericardial injection of gelfoam was safe and reliable. Presence of the MSCs and eGFP expression from adenovirus in the myocardium were confirmed after delivery. Our novel percutaneous approach to deliver (stem-) cells or adenovirus was safe and efficient in this pre-clinical model. IVUS-guided delivery is a minimally invasive procedure that seems to be a promising new strategy to deliver therapeutic agents locally to the heart.


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.

Collaboration


Dive into the Kiyotake Ishikawa's collaboration.

Top Co-Authors

Avatar

Roger J. Hajjar

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

Lisa Tilemann

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
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge