Network


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

Hotspot


Dive into the research topics where Yunzeng Zou is active.

Publication


Featured researches published by Yunzeng Zou.


Journal of Clinical Investigation | 1997

Oxidative stress activates extracellular signal-regulated kinases through Src and Ras in cultured cardiac myocytes of neonatal rats.

Ryuichi Aikawa; Issei Komuro; Tsutomu Yamazaki; Yunzeng Zou; Sumiyo Kudoh; Mariko Tanaka; Ichiro Shiojima; Yukio Hiroi; Yoshio Yazaki

A growing body of evidence has suggested that oxidative stress causes cardiac injuries during ischemia/reperfusion. Extracellular signal-regulated kinases (ERKs) have been reported to play pivotal roles in many aspects of cell functions and to be activated by oxidative stress in some types of cells. In this study, we examined oxidative stress-evoked signal transduction pathways leading to activation of ERKs in cultured cardiomyocytes of neonatal rats, and determined their role in oxidative stress-induced cardiomyocyte injuries. ERKs were transiently and concentration-dependently activated by hydrogen peroxide (H2O2) in cardiac myocytes. A specific tyrosine kinase inhibitor, genistein, suppressed H2O2-induced ERK activation, while inhibitors of protein kinase A and C or Ca2+ chelators had no effects on the activation. When CSK, a negative regulator of Src family tyrosine kinases, or dominant-negative mutant of Ras or of Raf-1 kinase was overexpressed, activation of transfected ERK2 by H2O2 was abolished. The treatment with H2O2 increased the number of cells stained positive by terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling, and induced formation of DNA ladder and activation of CPP32, suggesting that H2O2 induced apoptosis of cardiac myocytes. When H2O2-induced activation of ERKs was selectively inhibited by PD98059, the number of cardiac myocytes which showed apoptotic death was increased. These results suggest that Src family tyrosine kinases, Ras and Raf-1 are critical for ERK activation by hydroxyl radicals and that activation of ERKs may play an important role in protecting cardiac myocytes from apoptotic death following oxidative stress.


Nature | 2007

p53-induced inhibition of Hif-1 causes cardiac dysfunction during pressure overload

Masanori Sano; Tohru Minamino; Haruhiro Toko; Hideyuki Miyauchi; Masayuki Orimo; Yingjie Qin; Hiroshi Akazawa; Kaoru Tateno; Yosuke Kayama; Mutsuo Harada; Ippei Shimizu; Takayuki Asahara; Hirofumi Hamada; Shuhei Tomita; Jeffrey D. Molkentin; Yunzeng Zou; Issei Komuro

Cardiac hypertrophy occurs as an adaptive response to increased workload to maintain cardiac function. However, prolonged cardiac hypertrophy causes heart failure, and its mechanisms are largely unknown. Here we show that cardiac angiogenesis is crucially involved in the adaptive mechanism of cardiac hypertrophy and that p53 accumulation is essential for the transition from cardiac hypertrophy to heart failure. Pressure overload initially promoted vascular growth in the heart by hypoxia-inducible factor-1 (Hif-1)-dependent induction of angiogenic factors, and inhibition of angiogenesis prevented the development of cardiac hypertrophy and induced systolic dysfunction. Sustained pressure overload induced an accumulation of p53 that inhibited Hif-1 activity and thereby impaired cardiac angiogenesis and systolic function. Conversely, promoting cardiac angiogenesis by introducing angiogenic factors or by inhibiting p53 accumulation developed hypertrophy further and restored cardiac dysfunction under chronic pressure overload. These results indicate that the anti-angiogenic property of p53 may have a crucial function in the transition from cardiac hypertrophy to heart failure.


Nature Medicine | 2005

G-CSF prevents cardiac remodeling after myocardial infarction by activating the Jak-Stat pathway in cardiomyocytes

Mutsuo Harada; Yingjie Qin; Hiroyuki Takano; Tohru Minamino; Yunzeng Zou; Haruhiro Toko; Masashi Ohtsuka; Katsuhisa Matsuura; Masanori Sano; Jun-ichiro Nishi; Koji Iwanaga; Hiroshi Akazawa; Takeshige Kunieda; Weidong Zhu; Hiroshi Hasegawa; Keita Kunisada; Toshio Nagai; Haruaki Nakaya; Keiko Yamauchi-Takihara; Issei Komuro

Granulocyte colony-stimulating factor (G-CSF) was reported to induce myocardial regeneration by promoting mobilization of bone marrow stem cells to the injured heart after myocardial infarction, but the precise mechanisms of the beneficial effects of G-CSF are not fully understood. Here we show that G-CSF acts directly on cardiomyocytes and promotes their survival after myocardial infarction. G-CSF receptor was expressed on cardiomyocytes and G-CSF activated the Jak/Stat pathway in cardiomyocytes. The G-CSF treatment did not affect initial infarct size at 3 d but improved cardiac function as early as 1 week after myocardial infarction. Moreover, the beneficial effects of G-CSF on cardiac function were reduced by delayed start of the treatment. G-CSF induced antiapoptotic proteins and inhibited apoptotic death of cardiomyocytes in the infarcted hearts. G-CSF also reduced apoptosis of endothelial cells and increased vascularization in the infarcted hearts, further protecting against ischemic injury. All these effects of G-CSF on infarcted hearts were abolished by overexpression of a dominant-negative mutant Stat3 protein in cardiomyocytes. These results suggest that G-CSF promotes survival of cardiac myocytes and prevents left ventricular remodeling after myocardial infarction through the functional communication between cardiomyocytes and noncardiomyocytes.


Nature Cell Biology | 2004

Mechanical stress activates angiotensin II type 1 receptor without the involvement of angiotensin II

Yunzeng Zou; Hiroshi Akazawa; Yingjie Qin; Masanori Sano; Hiroyuki Takano; Tohru Minamino; Noriko Makita; Koji Iwanaga; Weidong Zhu; Sumiyo Kudoh; Haruhiro Toko; Koichi Tamura; Minoru Kihara; Toshio Nagai; Akiyoshi Fukamizu; Satoshi Umemura; Taroh Iiri; Toshiro Fujita; Issei Komuro

The angiotensin II type 1 (AT1) receptor has a crucial role in load-induced cardiac hypertrophy. Here we show that the AT1 receptor can be activated by mechanical stress through an angiotensin-II-independent mechanism. Without the involvement of angiotensin II, mechanical stress not only activates extracellular-signal-regulated kinases and increases phosphoinositide production in vitro, but also induces cardiac hypertrophy in vivo. Mechanical stretch induces association of the AT1 receptor with Janus kinase 2, and translocation of G proteins into the cytosol. All of these events are inhibited by the AT1 receptor blocker candesartan. Thus, mechanical stress activates AT1 receptor independently of angiotensin II, and this activation can be inhibited by an inverse agonist of the AT1 receptor.


Journal of Biological Chemistry | 1996

Endothelin-1 Is Involved in Mechanical Stress-induced Cardiomyocyte Hypertrophy

Tsutomu Yamazaki; Issei Komuro; Sumiyo Kudoh; Yunzeng Zou; Ichiro Shiojima; Yukio Hiroi; Takehiko Mizuno; Koji Maemura; Hiroki Kurihara; Ryuichi Aikawa; Hiroyuki Takano; Yoshio Yazaki

We have recently shown that mechanical stress induces cardiomyocyte hypertrophy partly through the enhanced secretion of angiotensin II (ATII). Endothelin-1 (ET-1) has been reported to be a potent growth factor for a variety of cells, including cardiomyocytes. In this study, we examined the role of ET-1 in mechanical stress-induced cardiac hypertrophy by using cultured cardiomyocytes of neonatal rats. ET-1 (1010M) maximally induced the activation of both Raf-1 kinase and mitogen-activated protein (MAP) kinases at 4 and 8 min, respectively, followed by an increase in protein synthesis at 24 h. All of these hypertrophic responses were completely blocked by pretreatment with BQ123, an antagonist selective for the ET-1 type A receptor subtype, but not by BQ788, an ET-1 type B receptor-specific antagonist. BQ123 also suppressed stretch-induced activation of MAP kinases and an increase in phenylalanine uptake by approximately 60 and 50%, respectively, but BQ788 did not. ET-1 was constitutively secreted from cultured cardiomyocytes, and a significant increase in ET-1 concentration was observed in the culture medium of cardiomyocytes after stretching for 10 min. After 24 h, an 3-fold increase in ET-1 concentration was observed in the conditioned medium of stretched cardiomyocytes compared with that of unstretched cardiomyocytes. ET-1 mRNA levels were also increased at 30 min after stretching. Moreover, ET-1 and ATII synergistically activated Raf-1 kinase and MAP kinases in cultured cardiomyocytes. In conclusion, mechanical stretching stimulates secretion and production of ET-1 in cultured cardiomyocytes, and vasoconstrictive peptides such as ATII and ET-1 may play an important role in mechanical stress-induced cardiac hypertrophy.


Circulation Research | 1995

Angiotensin II partly mediates mechanical stress-induced cardiac hypertrophy.

Tsutomu Yamazaki; Issei Komuro; Sumiyo Kudoh; Yunzeng Zou; Ichiro Shiojima; Takehiko Mizuno; Hiroyuki Takano; Yukio Hiroi; Kohjiro Ueki; Kazuyuki Tobe; Takashi Kadowaki; Ryozo Nagai; Yoshio Yazaki

We have previously shown that mechanical stress induces activation of protein kinases and increases in specific gene expression and protein synthesis in cardiac myocytes, all of which are similar to those evoked by humoral factors such as growth factors and hormones. Many lines of evidence have suggested that angiotensin II (Ang II) plays a vital role in cardiac hypertrophy, and it has been reported that secretion of Ang II from cultured cardiac myocytes was induced by mechanical stretch. To examine the role of Ang II in mechanical stress-induced cardiac hypertrophy, we stretched neonatal rat cardiac myocytes in the absence or presence of the Ang II receptor antagonists saralasin (an antagonist of both type 1 and type 2 receptors), CV-11974 (a type 1 receptor-specific antagonist), and PD123319 (a type 2 receptor-specific antagonist). Stretching cardiac myocytes by 20% using deformable silicone dishes rapidly increased the activities of mitogen-activated protein (MAP) kinase kinase activators and MAP kinases. Both saralasin and CV-11974 partially inhibited the stretch-induced increases in the activities of both kinases, whereas PD123319 showed no inhibitory effects. Stretching cardiac myocytes increased amino acid incorporation, which was also inhibited by approximately 70% with the pretreatment by saralasin or CV-11974. When the culture medium conditioned by stretching cardiocytes was transferred to nonstretched cardiac myocytes, the increase in MAP kinase activity was observed, and this increase was completely suppressed by saralasin or CV-11974. These results suggest that Ang II plays an important role in mechanical stress-induced cardiac hypertrophy and that there are also other (possibly nonsecretory) factors to induce hypertrophic responses.


The FASEB Journal | 1996

Mechanical stretch activates the stress-activated protein kinases in cardiac myocytes.

Issei Komuro; S Kudo; Tsutomu Yamazaki; Yunzeng Zou; Ichiro Shiojima; Yoshio Yazaki

We have recently shown that mechanical stress activates a phosphorylation cascade of protein kinases including Raf‐1 and the extracellular signal‐regulated kinases (ERKs) in cultured cardiac myocytes partially through the enhanced secretion of angiotensin II. Osmotic stress in budding yeast has been shown to activate similar signaling molecules including Hog‐1, a distant relative of the ERK family. In the present study, we examined whether mechanical stretch of cardiac myocytes activates the stress‐activated protein kinases (SAPKs)/c‐Jun NH2‐terminal kinase, the mammalian homologs of yeast Hog‐1 that regulate gene expression through activation of the transcription factor, AP‐l. When cardiac myocytes of neonatal rats cultured on a deformable silicone dish were stretched, activity of SAPKs was increased from 10 min, peaked at 30 min, and gradually decreased thereafter. The increase in activity of SAPKs was proportional to the stretch. Unlike ERKs, the activation of SAPKs by stretching cardiac myocytes was not dependent on the secreted angiotensin II. The chelation of extracellular Ca2+ or down‐regulation of protein kinase C did not attenuate activation of SAPKs by stretch. Transfection experiments using an AP‐l binding site‐containing reporter gene revealed that stretch increases AP‐l activity in cardiac myocytes. In conclusion, like osmotic stress in yeast, mechanical stretch activates SAPKs in cardiac myocytes without the participation of angiotensin II. These results suggest that the activation of SAPKs may regulate gene expression during mechanical stress‐induced cardiac hypertrophy.—Komuro, I., Kudo, S., Yamazaki, T., Zou, Y., Shiojima, I., Yazaki, Y. Mechanical stretch activates the stress‐activated protein kinases in cardiac myocytes. FASEB J. 10, 631‐636 (1996)


Basic Research in Cardiology | 2005

Time course of myocardial stromal cell–derived factor 1 expression and beneficial effects of intravenously administered bone marrow stem cells in rats with experimental myocardial infarction

Jun Ma; Junbo Ge; Shaoheng Zhang; Aijun Sun; Jianying Shen; Leilei Chen; Keqiang Wang; Yunzeng Zou

ObjectiveThe chemokine stromal cell–derived factor–1 (SDF–1) has been implicated in homing of bone marrow cells to sites of injury. We investigated the time course of myocardial SDF–1 expression and effects of intravenously administered bone marrow mesenchymal stem cells (MSC) in rats with myocardial infarction (MI).MethodsSDF–1 expression was measured by RT–PCR and Western blot in sham operated or infarcted hearts at 1/2, 1, 2, 4, 8 and 16 days post operation. MSCs from donor rats were labeled with BrdU. A total of 5 × 106 cells in 2.5 mL of PBS or equal volume PBS alone were injected through the tail vein at above mentioned time points. The number of the labeled MSCs in the infarcted hearts was counted 3 days post injection. Cardiac function and vessel numbers were assessed 28 days post injection.ResultsMyocardial SDF–1 expression increased and peaked at the first day and decreased thereafter post MI and remained unchanged in sham operated hearts. The MSCs enrichment and angiogenesis in the host hearts were more abundant in the 1 day transplantation group than in the other groups (P < 0.01). Cardiac function was only improved in rats received intravenous MSCs injection within 4 days post MI and not affected by PBS injection.Conclusions Myocardial SDF–1 expression was increased only in the early phase post MI. MSCs intravenous infused at the early phase of MI were recruited to injured heart, enhanced angiogenesis and improved cardiac function.


Journal of Biological Chemistry | 1996

Protein Kinase C, but Not Tyrosine Kinases or Ras, Plays a Critical Role in Angiotensin II-induced Activation of Raf-1 Kinase and Extracellular Signal-regulated Protein Kinases in Cardiac Myocytes

Yunzeng Zou; Issei Komuro; Tsutomu Yamazaki; Ryuichi Aikawa; Sumiyo Kudoh; Ichiro Shiojima; Yukio Hiroi; Takehiko Mizuno; Yoshio Yazaki

Angiotensin II (AngII) induces cardiac hypertrophy through activating a variety of protein kinases. In this study, to understand how cardiac hypertrophy develops, we examined AngII-evoked signal transduction pathways leading to the activation of extracellular signal-regulated protein kinases (ERKs), which are reportedly critical for the development of cardiac hypertrophy, in cultured cardiac myocytes isolated from neonatal rats. Inhibition of protein kinase C (PKC) with calphostin C or down-regulation of PKC by pretreatment with a phorbol ester for 24 h abolished AngII-induced activation of Raf-1 and ERKs, and addition of a phorbol ester conversely induced a marked increase in the activities of Raf-1 and ERKs. Pretreatment with two chemically and mechanistically dissimilar tyrosine kinase inhibitors, genistein and tyrphostin, did not attenuate AngII-induced activation of ERKs. In contrast, genistein strongly blocked insulin-induced ERK activation in cardiac myocytes. Although pretreatment with manumycin, a Ras farnesyltransferase inhibitor, or overexpression of a dominant-negative mutant of Ras inhibited insulin-induced ERK activation, neither affected AngII-induced activation of ERKs. Overexpression of a dominant-negative mutant of Raf-1 completely suppressed ERK2 activation by AngII, endothelin-1, and insulin. These results suggest that PKC and Raf-1, but not tyrosine kinases or Ras, are critical for AngII-induced activation of ERKs in cardiac myocytes.


The FASEB Journal | 2004

Cytokine therapy prevents left ventricular remodeling and dysfunction after myocardial infarction through neovascularization

Masashi Ohtsuka; Hiroyuki Takano; Yunzeng Zou; Haruhiro Toko; Hiroshi Akazawa; Yingjie Qin; Masashi Suzuki; Hiroshi Hasegawa; Haruaki Nakaya; Issei Komuro

Pretreatment with a combination of granulocyte colony‐stimulating factor (G‐CSF) and stem cell factor (SCF) has been reported to attenuate left ventricular (LV) remodeling after acute myocardial infarction (MI). We here examined whether the cytokine treatment started after MI has also beneficial effects. Anterior MI was created in the recipient mice whose bone marrow had been replaced with that of transgenic mice expressing enhanced green fluorescent protein (GFP). We categorized mice into five groups according to the following treatment: 1) saline; 2) administration of G‐CSF and SCF from 5 days before MI through 3 days after; 3) administration of G‐CSF and SCF for 5 days after MI; 4) administration of G‐CSF alone for 5 days after MI; 5) administration of SCF alone for 5 days after MI. All the three treatment groups with G‐CSF showed less LV remodeling and improved cardiac function and survival rate after MI. The number of capillaries, which express GFP, was increased and the number of apoptotic cells was decreased in the border area of all the treatment groups with G‐CSF. Even if the cytokine treatment is started after MI, it could prevent LV remodeling and dysfunction after MI—at least in part—through an increase in neovascularization and a decrease in apoptosis in the border area.

Collaboration


Dive into the Yunzeng Zou'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