Network


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

Hotspot


Dive into the research topics where Ang Guo is active.

Publication


Featured researches published by Ang Guo.


Circulation Research | 2010

T-Tubule Remodeling During Transition From Hypertrophy to Heart Failure

Sheng Wei; Ang Guo; Biyi Chen; William Kutschke; Yu-Ping Xie; Kathy Zimmerman; Robert M. Weiss; Mark E. Anderson; Heping Cheng; Long-Sheng Song

Rationale: The transverse tubule (T-tubule) system is the ultrastructural substrate for excitation–contraction coupling in ventricular myocytes; T-tubule disorganization and loss are linked to decreased contractility in end stage heart failure (HF). Objective: We sought to examine (1) whether pathological T-tubule remodeling occurs early in compensated hypertrophy and, if so, how it evolves during the transition from hypertrophy to HF; and (2) the role of junctophilin-2 in T-tubule remodeling. Methods and Results: We investigated T-tubule remodeling in relation to ventricular function during HF progression using state-of-the-art confocal imaging of T-tubules in intact hearts, using a thoracic aortic banding rat HF model. We developed a quantitative T-tubule power (TTpower) index to represent the integrity of T-tubule structure. We found that discrete local loss and global reorganization of the T-tubule system (leftward shift of TTpower histogram) started early in compensated hypertrophy in left ventricular (LV) myocytes, before LV dysfunction, as detected by echocardiography. With progression from compensated hypertrophy to early and late HF, T-tubule remodeling spread from the LV to the right ventricle, and TTpower histograms of both ventricles gradually shifted leftward. The mean LV TTpower showed a strong correlation with ejection fraction and heart weight to body weight ratio. Over the progression to HF, we observed a gradual reduction in the expression of a junctophilin protein (JP-2) implicated in the formation of T-tubule/sarcoplasmic reticulum junctions. Furthermore, we found that JP-2 knockdown by gene silencing reduced T-tubule structure integrity in cultured adult ventricular myocytes. Conclusions: T-tubule remodeling in response to thoracic aortic banding stress begins before echocardiographically detectable LV dysfunction and progresses over the development of overt structural heart disease. LV T-tubule remodeling is closely associated with the severity of cardiac hypertrophy and predicts LV function. Thus, T-tubule remodeling may constitute a key mechanism underlying the transition from compensated hypertrophy to HF.


Nature Medicine | 2011

Carvedilol and its new analogs suppress arrhythmogenic store overload-induced Ca2+ release

Qiang Zhou; Jianmin Xiao; Dawei Jiang; Ruiwu Wang; Kannan Vembaiyan; Aixia Wang; Christopher Smith; Cuihong Xie; Wenqian Chen; Jingqun Zhang; Xixi Tian; Peter P. Jones; Xiaowei Zhong; Ang Guo; Haiyan Chen; Lin Zhang; Weizhong Zhu; Dongmei Yang; Xiaodong Li; Ju Chen; Anne M. Gillis; Henry J. Duff; Heping Cheng; Arthur M. Feldman; Long-Sheng Song; Michael Fill; Thomas G. Back; S. R. Wayne Chen

Carvedilol is one of the most effective beta blockers for preventing ventricular tachyarrhythmias in heart failure, but the mechanisms underlying its favorable antiarrhythmic benefits remain unclear. Spontaneous Ca2+ waves, also called store overload–induced Ca2+ release (SOICR), evoke ventricular tachyarrhythmias in individuals with heart failure. Here we show that carvedilol is the only beta blocker tested that effectively suppresses SOICR by directly reducing the open duration of the cardiac ryanodine receptor (RyR2). This unique anti-SOICR activity of carvedilol, combined with its beta-blocking activity, probably contributes to its favorable antiarrhythmic effect. To enable optimal titration of carvedilols actions as a beta blocker and as a suppressor of SOICR separately, we developed a new SOICR-inhibiting, minimally beta-blocking carvedilol analog, VK-II-86. VK-II-86 prevented stress-induced ventricular tachyarrhythmias in RyR2-mutant mice and did so more effectively when combined with either of the selective beta blockers metoprolol or bisoprolol. Combining SOICR inhibition with optimal beta blockade has the potential to provide antiarrhythmic therapy that can be tailored to individual patients.


Nature Medicine | 2014

The ryanodine receptor store-sensing gate controls Ca2+ waves and Ca2+-triggered arrhythmias

Wenqian Chen; Ruiwu Wang; Biyi Chen; Xiaowei Zhong; Huihui Kong; Yunlong Bai; Qiang Zhou; Cuihong Xie; Jingqun Zhang; Ang Guo; Xixi Tian; Peter P. Jones; Megan L. O'Mara; Yingjie Liu; Tao Mi; Lin Zhang; Jeff Bolstad; Lisa Semeniuk; Hongqiang Cheng; Jianlin Zhang; Ju Chen; D. Peter Tieleman; Anne M. Gillis; Henry J. Duff; Michael Fill; Long-Sheng Song; S. R. Wayne Chen

Spontaneous Ca2+ release from intracellular stores is important for various physiological and pathological processes. In cardiac muscle cells, spontaneous store overload–induced Ca2+ release (SOICR) can result in Ca2+ waves, a major cause of ventricular tachyarrhythmias (VTs) and sudden death. The molecular mechanism underlying SOICR has been a mystery for decades. Here we show that a point mutation, E4872A, in the helix bundle crossing region (the proposed gate) of the cardiac ryanodine receptor (RyR2) completely abolishes luminal, but not cytosolic, Ca2+ activation of RyR2. The introduction of metal-binding histidines at this site converts RyR2 into a luminal Ni2+-gated channel. Mouse hearts harboring a heterozygous RyR2 mutation at this site (E4872Q) are resistant to SOICR and are completely protected against Ca2+-triggered VTs. These data show that the RyR2 gate directly senses luminal (store) Ca2+, explaining the regulation of RyR2 by luminal Ca2+, the initiation of Ca2+ waves and Ca2+-triggered arrhythmias. This newly identified store-sensing gate structure is conserved in all RyR and inositol 1,4,5-trisphosphate receptor isoforms.


Cardiovascular Research | 2013

Emerging mechanisms of T-tubule remodelling in heart failure

Ang Guo; Caimei Zhang; Sheng Wei; Biyi Chen; Long-Sheng Song

Cardiac excitation-contraction coupling occurs primarily at the sites of transverse (T)-tubule/sarcoplasmic reticulum junctions. The orderly T-tubule network guarantees the instantaneous excitation and synchronous activation of nearly all Ca(2+) release sites throughout the large ventricular myocyte. Because of the critical roles played by T-tubules and the array of channels and transporters localized to the T-tubule membrane network, T-tubule architecture has recently become an area of considerable research interest in the cardiovascular field. This review will focus on the current knowledge regarding normal T-tubule structure and function in the heart, T-tubule remodelling in the transition from compensated hypertrophy to heart failure, and the impact of T-tubule remodelling on myocyte Ca(2+) handling function. In the last section, we discuss the molecular mechanisms underlying T-tubule remodelling in heart disease.


Hypertension | 2012

Sildenafil prevents and reverses transverse-tubule remodeling and Ca(2+) handling dysfunction in right ventricle failure induced by pulmonary artery hypertension.

Yu Ping Xie; Biyi Chen; Philip N. Sanders; Ang Guo; Yue Li; Kathy Zimmerman; Lie Cheng Wang; Robert M. Weiss; Isabella M. Grumbach; Mark E. Anderson; Long-Sheng Song

Right ventricular (RV) failure (RVF) is the main cause of death in patients with pulmonary artery hypertension (PAH). Sildenafil, a phosphodiesterase type 5 inhibitor, was approved recently for treatment of PAH patients. However, the mechanisms underlying RV contractile malfunction and the benefits of sildenafil on RV function are not well understood. We aimed to investigate the following: (1) the ultrastructural and excitation-contraction coupling alterations underlying PAH-induced RVF; (2) whether the ultrastructural changes are reversible; and (3) the mechanisms underlying the therapeutic benefits of sildenafil in PAH-RVF. We used a single injection of monocrotaline in Wistar rats to induce pulmonary vascular proliferation, which led to PAH and RVF. RV myocytes displayed severe transverse (T)-tubule loss and disorganization, as well as blunted and dys-synchronous sarcoplasmic reticulum Ca2+ release. Sildenafil prevented and reversed the monocrotaline-induced PAH and LV filling impairment. Early intervention with sildenafil prevented RV hypertrophy and the development of RVF, T-tubule remodeling, and Ca2+ handling dysfunction. Although late treatment with sildenafil did not reverse RV hypertrophy in animals with established RVF, RV systolic function was improved. Furthermore, late intervention partially reversed both the impairment of myocyte T-tubule integrity and Ca2+ handling protein and sarcoplasmic reticulum Ca2+ release function in monocrotaline-treated rats. In conclusion, PAH-induced increase in RV afterload causes severe T-tubule remodeling and Ca2+ handling dysfunction in RV myocytes, leading to RV contractile failure. Sildenafil prevents and partially reverses ultrastructural, molecular, and functional remodeling of failing RV myocytes. Reversal of pathological T-tubule remodeling, although incomplete, is achievable without the regression of RV hypertrophy.


Circulation | 2014

Microtubule-Mediated Defects in Junctophilin-2 Trafficking Contribute to Myocyte Transverse-Tubule Remodeling and Ca2+ Handling Dysfunction in Heart Failure

Caimei Zhang; Biyi Chen; Ang Guo; Yanqi Zhu; Jordan D. Miller; Shan Gao; Can Yuan; William Kutschke; Kathy Zimmerman; Robert M. Weiss; Xander H.T. Wehrens; Jiang Hong; Frances L. Johnson; Luis F. Santana; Mark E. Anderson; Long-Sheng Song

Background— Cardiac dysfunction in failing hearts of human patients and animal models is associated with both microtubule densification and transverse-tubule (T-tubule) remodeling. Our objective was to investigate whether microtubule densification contributes to T-tubule remodeling and excitation–contraction coupling dysfunction in heart disease. Methods and Results— In a mouse model of pressure overload–induced cardiomyopathy by transaortic banding, colchicine, a microtubule depolymerizer, significantly ameliorated T-tubule remodeling and cardiac dysfunction. In cultured cardiomyocytes, microtubule depolymerization with nocodazole or colchicine profoundly attenuated T-tubule impairment, whereas microtubule polymerization/stabilization with taxol accelerated T-tubule remodeling. In situ immunofluorescence of heart tissue sections demonstrated significant disorganization of junctophilin-2 (JP2), a protein that bridges the T-tubule and sarcoplasmic reticulum membranes, in transaortic banded hearts as well as in human failing hearts, whereas colchicine injection significantly preserved the distribution of JP2 in transaortic banded hearts. In isolated mouse cardiomyocytes, prolonged culture or treatment with taxol resulted in pronounced redistribution of JP2 from T-tubules to the peripheral plasma membrane, without changing total JP2 expression. Nocodazole treatment antagonized JP2 redistribution. Moreover, overexpression of a dominant-negative mutant of kinesin 1, a microtubule motor protein responsible for anterograde trafficking of proteins, protected against JP2 redistribution and T-tubule remodeling in culture. Finally, nocodazole treatment improved Ca2+ handling in cultured myocytes by increasing the amplitude of Ca2+ transients and reducing the frequency of Ca2+ sparks. Conclusion— Our data identify a mechanistic link between microtubule densification and T-tubule remodeling and reveal microtubule-mediated JP2 redistribution as a novel mechanism for T-tubule disruption, loss of excitation–contraction coupling, and heart failure.


Proceedings of the National Academy of Sciences of the United States of America | 2014

Overexpression of junctophilin-2 does not enhance baseline function but attenuates heart failure development after cardiac stress

Ang Guo; Xiaoying Zhang; Venkat Ramesh Iyer; Biyi Chen; Caimei Zhang; William Kutschke; Robert M. Weiss; Clara Franzini-Armstrong; Long-Sheng Song

Significance Normal cardiac function requires coordinated contraction of working myocytes, which is initiated by a specific communication between Ca2+ channels on the transverse (T)-tubule membrane and ryanodine receptors on the sarcoplasmic reticulum (SR) membrane. Junctophilin-2 (JP2) is a structural protein that induces docking of SR to T-tubules to form dyads and that indirectly stabilizes the T-tubule network in ventricular cardiomyocytes. JP2 is frequently down-regulated in heart failure, in parallel with a disruption of the T-tubule network and loss of normal excitation-contraction coupling. Here we show that overexpression of JP2 stabilizes the T-tubule network and attenuates heart failure after cardiac stress. These data suggest that future treatment of heart disease may include strategies to stabilize the architecture of T-tubules and cardiac dyads. Heart failure is accompanied by a loss of the orderly disposition of transverse (T)-tubules and a decrease of their associations with the junctional sarcoplasmic reticulum (jSR). Junctophilin-2 (JP2) is a structural protein responsible for jSR/T-tubule docking. Animal models of cardiac stresses demonstrate that down-regulation of JP2 contributes to T-tubule disorganization, loss of excitation-contraction coupling, and heart failure development. Our objective was to determine whether JP2 overexpression attenuates stress-induced T-tubule disorganization and protects against heart failure progression. We therefore generated transgenic mice with cardiac-specific JP2 overexpression (JP2-OE). Baseline cardiac function and Ca2+ handling properties were similar between JP2-OE and control mice. However, JP2-OE mice displayed a significant increase in the junctional coupling area between T-tubules and the SR and an elevated expression of the Na+/Ca2+ exchanger, although other excitation-contraction coupling protein levels were not significantly changed. Despite similar cardiac function at baseline, overexpression of JP2 provided significantly protective benefits after pressure overload. This was accompanied by a decreased percentage of surviving mice that developed heart failure, as well as preservation of T-tubule network integrity in both the left and right ventricles. Taken together, these data suggest that strategies to maintain JP2 levels can prevent the progression from hypertrophy to heart failure.


The FASEB Journal | 2012

β-Adrenergic receptor antagonists ameliorate myocyte T-tubule remodeling following myocardial infarction

Biyi Chen; Yue Li; Shuxia Jiang; Yu Ping Xie; Ang Guo; William Kutschke; Kathy Zimmerman; Robert M. Weiss; Francis J. Miller; Mark E. Anderson; Long-Sheng Song

β‐Adrenergic receptor (AR) blockers provide substantial clinical benefits, including improving overall survival and left ventricular (LV) function following myocardial infarction (MI), though the mechanisms remain incompletely defined. The transverse‐tubule (T‐tubule) system of ventricular myocytes is an important determinant of cardiac excitation‐contraction function. T‐tubule remodeling occurs early during LV failure. We hypothesized that β‐AR blockers prevent T‐tubule remodeling and thereby provide therapeutic benefits. A murine model of MI was utilized to examine the effect of β‐AR blockers on T‐tubule remodeling following LV MI. We applied the in situ imaging of T‐tubule structure from Langendorff‐perfused intact hearts with laser scanning confocal microscopy. We found that MI caused remarkable T‐tubule remodeling near the infarction border zone and moderate LV remodeling remote from the MI. Metoprolol and carvedilol administered 6 d after MI for 4 wk each increased the T‐tubule integrity at the remote and border zones. At the molecular level, both β‐AR blockers restored border and remote zone expression of junctophilin‐2 (JP‐2), which is involved in T‐tubule organization and formation of the T‐tubule/sarcoplasmic reticulum junctions. In contrast, β‐AR blockers had no significant effects on caveolin‐3 expression. In summary, our data show that β‐AR antagonists can protect against T‐tubule remodeling after MI, suggesting a novel therapeutic mechanism of action for this drug class. Preservation of JP‐2 expression may contribute to the beneficial effects of metoprolol and carvedilol on T‐tubule remodeling.— Chen, B., Li, Y., Jiang, S., Xie, Y.‐P., Guo, A., Kutschke, W., Zimmerman, K., Weiss, R. M., Miller, F. J., Anderson, M. E., Song, L.‐S. β‐Adrenergic receptor antagonists ameliorate myocyte T‐tubule remodeling following myocardial infarction. FASEB J. 26, 2531‐2537 (2012). www.fasebj.org


Cardiovascular Research | 2013

Critical roles of junctophilin-2 in T-tubule and excitation–contraction coupling maturation during postnatal development

Biyi Chen; Ang Guo; Caimei Zhang; Rong Chen; Yanqi Zhu; Jiang Hong; William Kutschke; Kathy Zimmerman; Robert M. Weiss; Leonid V. Zingman; Mark E. Anderson; Xander H.T. Wehrens; Long-Sheng Song

AIMS Emerging evidence indicates a critical role for junctophilin-2 (JP2) in T-tubule integrity and assembly of cardiac dyads in adult ventricular myocytes. In the postnatal stage, one of the critical features of myocyte maturation is development of the T-tubule system, though the mechanisms remain poorly understood. In this study, we aim to determine whether JP2 is required for normal cardiac T-tubule maturation. METHODS AND RESULTS Using in situ confocal imaging of intact murine hearts, we found T-tubules were absent in both left- and right-ventricular myocytes at postnatal Day 8 and did not appear until Day 10. Quantification of T-tubule structural integrity using the T-tubule power (TT(power)) index revealed a progressive increase in TT(power) between postnatal Days 10 and 19. By postnatal Day 19, TT(power) was similar to that in adult murine cardiomyocytes, indicative of a nearly matured T-tubule network. JP2 levels increased dramatically during development, reaching levels observed in adult hearts by postnatal Day 14. Deficiency of JP2, using a mouse model in which a JP2-specific shRNA is expressed during embryonic development, severely impaired T-tubule maturation, with equivalent decreases in the left- and right-ventricular TT(power). We also detected a gradual increase in the density of transverse but not longitudinal tubules during development, and JP2 deficiency abolished the increase in the density of transverse elements. Alterations in T-tubules caused significant reduction in Ca(2+) transient amplitude and marked increase in Ca(2+) release dyssynchrony, Ca(2+) alternans, and spontaneous Ca(2+) waves, leading to contractile failure. CONCLUSION Our data identify a critical role for JP2 in T-tubule and excitation-contraction coupling maturation during development.


Circulation Research | 2013

Phospholamban Knockout Breaks Arrhythmogenic Ca2+ Waves and Suppresses Catecholaminergic Polymorphic Ventricular Tachycardia in Mice

Yunlong Bai; Peter P. Jones; Jiqing Guo; Xiaowei Zhong; Robert B. Clark; Qiang Zhou; Ruiwu Wang; Alexander Vallmitjana; Raul Benitez; Leif Hove-Madsen; Lisa Semeniuk; Ang Guo; Long-Sheng Song; Henry J. Duff; S. R. Wayne Chen

Rationale: Phospholamban (PLN) is an inhibitor of cardiac sarco(endo)plasmic reticulum Ca2+ ATPase. PLN knockout (PLN-KO) enhances sarcoplasmic reticulum Ca2+ load and Ca2+ leak. Conversely, PLN-KO accelerates Ca2+ sequestration and aborts arrhythmogenic spontaneous Ca2+ waves (SCWs). An important question is whether these seemingly paradoxical effects of PLN-KO exacerbate or protect against Ca2+-triggered arrhythmias. Objective: We investigate the impact of PLN-KO on SCWs, triggered activities, and stress-induced ventricular tachyarrhythmias (VTs) in a mouse model of cardiac ryanodine-receptor (RyR2)-linked catecholaminergic polymorphic VT. Methods and Results: We generated a PLN-deficient, RyR2-mutant mouse model (PLN−/−/RyR2-R4496C+/−) by crossbreeding PLN-KO mice with catecholaminergic polymorphic VT–associated RyR2-R4496C mutant mice. Ca2+ imaging and patch-clamp recording revealed cell-wide propagating SCWs and triggered activities in RyR2-R4496C+/− ventricular myocytes during sarcoplasmic reticulum Ca2+ overload. PLN-KO fragmented these cell-wide SCWs into mini-waves and Ca2+ sparks and suppressed the triggered activities evoked by sarcoplasmic reticulum Ca2+ overload. Importantly, these effects of PLN-KO were reverted by partially inhibiting sarco(endo)plasmic reticulum Ca2+ ATPase with 2,5-di-tert-butylhydroquinone. However, Bay K, caffeine, or Li+ failed to convert mini-waves to cell-wide SCWs in PLN−/−/RyR2-R4496C+/− ventricular myocytes. Furthermore, ECG analysis showed that PLN-KO mice are not susceptible to stress-induced VTs. On the contrary, PLN-KO protected RyR2-R4496C mutant mice from stress-induced VTs. Conclusions: Our results demonstrate that despite severe sarcoplasmic reticulum Ca2+ leak, PLN-KO suppresses triggered activities and stress-induced VTs in a mouse model of catecholaminergic polymorphic VT. These data suggest that breaking up cell-wide propagating SCWs by enhancing Ca2+ sequestration represents an effective approach for suppressing Ca2+-triggered arrhythmias.

Collaboration


Dive into the Ang Guo's collaboration.

Top Co-Authors

Avatar

Long-Sheng Song

Roy J. and Lucille A. Carver College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Biyi Chen

Roy J. and Lucille A. Carver College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Caimei Zhang

Roy J. and Lucille A. Carver College of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jiang Hong

Shanghai Jiao Tong University

View shared research outputs
Top Co-Authors

Avatar

Kathy Zimmerman

Roy J. and Lucille A. Carver College of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yanqi Zhu

Shanghai Jiao Tong University

View shared research outputs
Top Co-Authors

Avatar

Jingqun Zhang

Rush University Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge