Network


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

Hotspot


Dive into the research topics where Junya Takagawa is active.

Publication


Featured researches published by Junya Takagawa.


Biochemical and Biophysical Research Communications | 2008

VEGF improves survival of mesenchymal stem cells in infarcted hearts

Jennifer Pons; Yu Huang; Janice Arakawa-Hoyt; Daniel Washko; Junya Takagawa; Jianqin Ye; William Grossman; Hua Su

Bone marrow-derived mesenchymal stem cells (MSC) are a promising source for cell-based treatment of myocardial infarction (MI), but existing strategies are restricted by low cell survival and engraftment. We examined whether vascular endothelial growth factor (VEGF) improve MSC viability in infarcted hearts. We found long-term culture increased MSC-cellular stress: expressing more cell cycle inhibitors, p16(INK), p21 and p19(ARF). VEGF treatment reduced cellular stress, increased pro-survival factors, phosphorylated-Akt and Bcl-xL expression and cell proliferation. Co-injection of MSCs with VEGF to MI hearts increased cell engraftment and resulted in better improvement of cardiac function than that injected with MSCs or VEGF alone. In conclusion, VEGF protects MSCs from culture-induce cellular stress and improves their viability in ischemic myocardium, which results in improvements of their therapeutic effect for the treatment of MI.


Molecular Therapy | 2009

Injection of bone marrow cell extract into infarcted hearts results in functional improvement comparable to intact cell therapy.

Yerem Yeghiazarians; Yan Zhang; Megha Prasad; Henry Shih; Shereen A. Saini; Junya Takagawa; Richard E. Sievers; Maelene L. Wong; Neel K. Kapasi; Rachel Mirsky; Juha W. Koskenvuo; Petros Minasi; Jianqin Ye; Mohan N. Viswanathan; Franca S. Angeli; Andrew J. Boyle; Matthew L. Springer; William Grossman

We compared therapeutic benefits of intramyocardial injection of unfractionated bone marrow cells (BMCs) versus BMC extract as treatments for myocardial infarction (MI), using closed-chest ultrasound-guided injection at a clinically relevant time post-MI. MI was induced in mice and the animals treated at day 3 with either: (i) BMCs from green fluorescent protein (GFP)-expressing mice (n = 14), (ii) BMC extract (n = 14), or (iii) saline control (n = 14). Six animals per group were used for histology at day 6 and the rest followed to day 28 for functional analysis. Ejection fraction was similarly improved in the BMC and extract groups versus control (40.6 +/- 3.4 and 39.1 +/- 2.9% versus 33.2 +/- 5.0%, P < 0.05) with smaller scar sizes. At day 6 but not day 28, both therapies led to significantly higher capillary area and number of arterioles versus control. At day 6, BMCs increased the number of cycling cardiomyocytes (CMs) versus control whereas extract therapy resulted in significant reduction in the number of apoptotic CMs at the border zone (BZ) versus control. Intracellular components within BMCs can enhance vascularity, reduce infarct size, improve cardiac function, and influence CM apoptosis and cycling early after therapy following MI. Intact cells are not necessary and death of implanted cells may be a major component of the benefit.


Science Translational Medicine | 2011

Donor Myocardial Infarction Impairs the Therapeutic Potential of Bone Marrow Cells by an Interleukin-1–Mediated Inflammatory Response

Xiaoyin Wang; Junya Takagawa; Viola Lam; Daniel J. Haddad; Diana Tobler; Pamela Y. Mok; Yan Zhang; Brian T. Clifford; Kranthi Pinnamaneni; Shereen A. Saini; Robert Su; Maya J. Bartel; Richard E. Sievers; Larry Carbone; Scott C. Kogan; Yerem Yeghiazarians; Michelle L. Hermiston; Matthew L. Springer

Inflammation after myocardial infarction may explain failure of bone marrow cells to improve cardiac function in infarcted recipient hearts. Getting to the Heart of Bone Marrow Therapy If arteries are the highways of the body, arterial blockage is like a jackknifed tractor-trailer: It blocks the flow of traffic and prevents passage. However, whereas a traffic accident may merely delay your commute, arterial blockage can result in the death of nutrient- and oxygen-starved cells that are no longer fed by the blood, such as happens with a myocardial infarction (MI). In rodent models, cells from the bone marrow can substantially improve cardiac function after MI, but attempts to translate these studies into humans have met with limited success. Now, Wang et al. show that this failure in humans may result from inflammation caused by the MI itself and that blocking inflammation with an inhibitor of the cytokine IL-1 restored the healing power of the bone marrow cells. The difference in outcomes observed in the two systems stemmed from the purity of the mouse system. Because genetically identical mice are readily available, healthy littermates supply more than enough donor bone marrow cells to successfully repair damaged heart tissue. Humans, however, are not quite so uniform. Donor cells had to come from the patients themselves in order to prevent rejection or graft-versus-host disease. Wang et al. hypothesized that bone marrow cells from MI patients would be different from those of healthy donors. To better mimic the human situation, the authors used donor bone marrow cells from mice that suffered MI and found that these cells were also hindered in their ability to repair cardiac function after MI. However, anti-inflammatory treatment of the donor, including inhibition IL-1, restored the ability of these cells to fix cardiac function. This reverse-translation study, which took a clinical observation and attempted to explain it in a rodent model, has not only provided insight into the local environment after MI; it also suggests a new option for successfully treating MI in the clinic. Preventing or reversing the proinflammatory change in the bone marrow cells may help them to cause a change of heart. Delivery of bone marrow cells (BMCs) to the heart has substantially improved cardiac function in most rodent models of myocardial infarction (MI), but clinical trials of BMC therapy have led to only modest improvements. Rodent models typically involve intramyocardial injection of BMCs from distinct donor individuals who are healthy. In contrast, autologous BMCs from individuals after MI are used for clinical trials. Using BMCs from donor mice after MI, we discovered that recent MI impaired BMC therapeutic efficacy. MI led to myocardial inflammation and an increased inflammatory state in the bone marrow, changing the BMC composition and reducing their efficacy. Injection of a general anti-inflammatory drug or a specific interleukin-1 inhibitor to donor mice after MI prevented this impairment. Our findings offer an explanation of why human trials have not matched the success of rodent experiments and suggest potential strategies to improve the success of clinical autologous BMC therapy.


Gene Therapy | 2006

AAV serotype-1 mediates early onset of gene expression in mouse hearts and results in better therapeutic effect

Hua Su; Yu Huang; Junya Takagawa; Alicia Bárcena; Janice Arakawa-Hoyt; Jianqin Ye; William Grossman; Yuet Wai Kan

Adeno-associated viral vectors (AAV) are attractive tool for gene therapy for coronary artery disease. However, gene expression in myocardium mediated by AAV serotype 2 (AAV2) does not peak until 4–6 weeks after gene transfer. This delayed gene expression may reduce its therapeutic potential for acute cardiac infarction. To determine whether earlier gene expression and better therapeutic effect could be achieved using a different serotype, CMV promoter driving the EPO gene (AAV-EPO) was packaged into AAV serotypes 1–5 capsids and injected into mouse myocardium. EPO expression was studied by measuring the hematocrits and EPO mRNA. After we found that AAV1 mediates the highest gene expression after 4 days of gene transduction, AAV-LacZ (CMV promoter driving LacZ gene expression) and MLCVEGF (hypoxia-inducible and cardiac-specific VEGF expression) were packaged into AAV1 and 2 capsids. LacZ expression was detected in AAV1-LacZ but not in AAV2-LacZ-injected hearts 1 day after vector injection. Compared to AAV2-MLCVEGF that mediated no significant VEGF expression, AAV1-MLCVEGF mediated 13.7-fold induction of VEGF expression in ischemic hearts 4 days after gene transduction and resulted in more neovasculatures, better cardiac function and less myocardial fibrosis. Thus, AAV1 mediates earlier and higher transgene expression in myocardium and better therapeutic effects.


Journal of Gene Medicine | 2009

Combining angiogenic gene and stem cell therapies for myocardial infarction

Jennifer Pons; Yu Huang; Junya Takagawa; Janice Arakawa-Hoyt; Jianqin Ye; William Grossman; Yuet Wai Kan; Hua Su

Transplantation of stem cells from various sources into infarcted hearts has the potential to promote myocardial regeneration. However, the regenerative capacity is limited partly as a result of the low survival rate of the transplanted cells in the ischemic myocardium. In the present study, we tested the hypothesis that combining cell and angiogenic gene therapies would provide additive therapeutic effects via co‐injection of bone marrow‐derived mesenchymal stem cells (MSCs) with an adeno‐associated viral vector (AAV), MLCVEGF, which expresses vascular endothelial growth factor (VEGF) in a cardiac‐specific and hypoxia‐inducible manner.


American Journal of Physiology-heart and Circulatory Physiology | 2014

Slow and deep respiration suppresses steady-state sympathetic nerve activity in patients with chronic heart failure: from modeling to clinical application

Daisuke Harada; Hidetsugu Asanoi; Junya Takagawa; Hisanari Ishise; Hiroshi Ueno; Yoshitaka Oda; Yukiko Goso; Shuji Joho; Hiroshi Inoue

Influences of slow and deep respiration on steady-state sympathetic nerve activity remain controversial in humans and could vary depending on disease conditions and basal sympathetic nerve activity. To elucidate the respiratory modulation of steady-state sympathetic nerve activity, we modeled the dynamic nature of the relationship between lung inflation and muscle sympathetic nerve activity (MSNA) in 11 heart failure patients with exaggerated sympathetic outflow at rest. An autoregressive exogenous input model was utilized to simulate entire responses of MSNA to variable respiratory patterns. In another 18 patients, we determined the influence of increasing tidal volume and slowing respiratory frequency on MSNA; 10 patients underwent a 15-min device-guided slow respiration and the remaining 8 had no respiratory modification. The model predicted that a 1-liter, step increase of lung volume decreased MSNA dynamically; its nadir (-33 ± 22%) occurred at 2.4 s; and steady-state decrease (-15 ± 5%), at 6 s. Actually, in patients with the device-guided slow and deep respiration, respiratory frequency effectively fell from 16.4 ± 3.9 to 6.7 ± 2.8/min (P < 0.0001) with a concomitant increase in tidal volume from 499 ± 206 to 1,177 ± 497 ml (P < 0.001). Consequently, steady-state MSNA was decreased by 31% (P < 0.005). In patients without respiratory modulation, there were no significant changes in respiratory frequency, tidal volume, and steady-state MSNA. Thus slow and deep respiration suppresses steady-state sympathetic nerve activity in patients with high levels of resting sympathetic tone as in heart failure.


Journal of the American College of Cardiology | 2002

Cardiac sympathetic denervationmodulates the sympathoexcitatoryresponse to acute myocardial ischemia

Shuji Joho; Hidetsugu Asanoi; Junya Takagawa; Tomoki Kameyama; Tadakazu Hirai; Takashi Nozawa; Katsumi Umeno; Masashi Shimizu; Hikaru Seto; Hiroshi Inoue

Abstract Objectives This study was designed to elucidate the influence of cardiac sympathetic denervation on the sympathoexcitatory response to acute myocardial ischemia during balloon coronary occlusion (BCO) in humans. Background Alterations of cardiac sympathetic nerve function could modulate sympathetic reflexes originating from the ischemic area. Methods In 23 patients with angina pectoris, we quantified the baseline cardiac sympathetic denervation of the ischemia-related area by iodine-123 metaiodobenzylguanidine (123I-MIBG), and transient changes in sympathetic activity during BCO by wavelet analysis of RR interval variability. Results Balloon coronary occlusion resulted in a transient augmentation of low-frequency (LF: 0.04 to 0.14 Hz) spectral components of RR interval variability in 4 of 12 patients with cardiac denervation and in 8 of 11 patients without denervation (p Conclusions These findings suggest that if the provoked ischemia is not severe, cardiac sympathetic denervation could prevent ischemia-induced sympathoexcitation.


Journal of Tissue Science and Engineering | 2011

Advanced Donor Age Impairs Bone Marrow Cell Therapeutic Efficacy for Cardiac Disease.

Xiaoyin Wang; Junya Takagawa; Daniel J. Haddad; Kranthi Pinnamaneni; Yan Zhang; Richard E. Sievers; William Grossman; Yerem Yeghiazarians; Matthew L. Springer

Therapeutic results of clinical autologous bone marrow cell (BMC) therapy trials for cardiac disease have been modest compared to results of BMC implantation into rodent hearts post-myocardial infarction (MI). In clinical trials, autologous BMCs are typically harvested from older patients who have recently suffered an MI. In contrast, experimental studies in rodent models typically utilize donor BMCs isolated from young, healthy, inbred mice that are not the recipients. Using unfractionated BMCs from donor mice at ages of young, middle-aged, and old, we discovered that recipient left ventricular function post-MI was significantly improved by young donor BMC implantation but was only preserved by middle-aged donor BMCs. Notably, old donor BMCs did not slow the decline in recipient post-MI cardiac function, suggesting BMC impairment by advanced donor age. Furthermore, we also show here that BMCs that are therapeutically impaired by donor age can be further impaired by concurrent donor MI. In conclusion, our findings suggest that therapeutic impairment of BMCs by advanced age is one of the important factors that can limit the success of clinical autologous BMC-based therapy.


Journal of Cardiology | 2017

Independent prognostic importance of respiratory instability and sympathetic nerve activity in patients with chronic heart failure

Hidetsugu Asanoi; Daisuke Harada; Yoshitaka Oda; Hiroshi Ueno; Junya Takagawa; Hisanari Ishise; Yukiko Goso; Shuji Joho; Hiroshi Inoue

BACKGROUND Respiratory instability in chronic heart failure (CHF) is characterized by irregularly rapid respiration or non-periodic breathing rather than by Cheyne-Stokes respiration. We developed a new quantitative measure of respiratory instability (RSI) and examined its independent prognostic impact upon CHF. METHODS In 87 patients with stable CHF, respiratory flow and muscle sympathetic nerve activity (MSNA) were simultaneously recorded. RSI was calculated from the frequency distribution of respiratory spectral components and very low frequency components. RESULTS During a mean follow-up of 85±38 months, 24 patients died. Sixteen patients who died of cardiac causes had a lower RSI (16±6 vs. 30±21, p<0.01), a lower specific activity scale (4.3±1.4 Mets vs. 5.7±1.4 Mets, p<0.005), a higher MSNA burst area (16±5% vs. 11±4%, p<0.001), and a higher brain natriuretic peptide (BNP) level (514±559pg/ml vs. 234±311pg/ml, p<0.05) than 71 patients who did not die of cardiac causes. Multivariate analysis revealed that RSI (p=0.015), followed by MSNA burst area (p=0.033), was an independent predictor of subsequent all-cause deaths and that RSI (p=0.026), MSNA burst area (p=0.001), and BNP (p=0.048) were independent predictors of cardiac deaths. Patients at very high risk of fatal outcome could be identified by an RSI<20. CONCLUSIONS The daytime respiratory instability quantified by a new measure of RSI has prognostic importance independent of sympathetic nerve activation in patients with clinically stable CHF. An RSI of <20 identifies patients at very high risk for subsequent all-cause and cardiovascular death.


Molecular Therapy | 2006

453. Injectionof Different Adult Cell Types into Mouse Myocardium Three Days after Myocardial Infarction Using a Novel Echo-Guided Technique Improves Cardiac Function

Yan Zhang; Junya Takagawa; Richard E. Sievers; Maelene L. Wong; Christian Heiss; Mohan N. Viswanathan; Neel K. Kapasi; Jianqin Ye; Elyse Foster; William Grossman; Matthew L. Springer; Yerem Yeghiazarians

Objective: Myocardial regeneration based on stem cell transplantation has emerged as a potential therapeutic approach toward replacing myocardial scar with functioning contractile tissue after myocardial infarction (MI). Considerable interest has focused on bone marrow cells (BMCs) and endothelial progenitor cells (EPCs) because they appear successful in attenuating remodeling following acute MI. However, preclinical experiments toward this goal have been limited to treatment within hours of an MI, in contrast to human trials, in which cell treatment has been performed several days later after patient stabilization and autologous cell harvesting. We sought to compare the therapeutic effects of different types of putative stem and progenitor cells in a mouse model of MI using a novel closed-chest echo-guided injection approach to deliver cells 3 days after infarction.

Collaboration


Dive into the Junya Takagawa's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge