Network


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

Hotspot


Dive into the research topics where Wenting Guo is active.

Publication


Featured researches published by Wenting Guo.


Science | 2016

Structural basis for the gating mechanism of the type 2 ryanodine receptor RyR2

Wei Peng; Huaizong Shen; Jianping Wu; Wenting Guo; Xiaojing Pan; Ruiwu Wang; S. R. Wayne Chen; Nieng Yan

Gating a calcium channel The type 2 ryanodine receptor (RyR2) controls the release of calcium ions from the sarcoplasmic reticulum in cardiac cells—the initiating step in cardiac muscle contraction. Mutations in RyR2 are associated with cardiac diseases. Peng et al. used single-particle electron cryomicroscopy to determine the structure of RyR2 from porcine heart at 4.4-Å resolution with the calcium channel closed and at 4.2-Å resolution with the calcium channel open. The structures reveal how interdomain motions result in a conformational change in the cytoplasmic region of RyR2 that is transduced by a central domain to cause motions that open or close the channel. Science, this issue p. 301 Motion of the cytoplasmic region of a key intracellular Ca2+ channel is transduced by a central domain to gate the channel domain. INTRODUCTION Ryanodine receptors (RyRs) are intracellular Ca2+ channels that control the release of Ca2+ from the sarco(endo)plasmic reticulum. Among the three mammalian RyR isoforms, RyR2 is primarily expressed in the heart and brain and is activated by Ca2+ influx by a mechanism known as calcium-induced calcium release. This Ca2+ release is fundamental to cellular processes ranging from muscle contraction to learning and memory. RyRs are the largest known ion channels with a molecular mass of > 2 megadalton for a homotetramer. Each RyR protomer consists of a cytoplasmic region of over 4500 residues and a carboxyl terminal transmembrane (TM) domain. The four identical TM segments enclose a central ion-conducting pore, whereas the cytoplasmic regions serve as a scaffold for interactions with diverse ligands and protein modulators. RyR channel gating involves a long-range allosteric mechanism. The structures of rabbit skeletal muscle RyR1 were determined at 3.8-Å resolution for the closed state and various lower resolutions for the potentially open states. Elucidating the gating mechanism of RyR requires structural determination of the open states at high resolution. RATIONALE RyR2 harbors more than 150 mutations associated with cardiac disorders, such as catecholaminergic polymorphic ventricular tachycardia type 1, idiopathic ventricular fibrillation, and sudden cardiac death. Structural elucidation of RyR2 may provide the molecular basis for understanding disease mechanisms and for the development of potential novel therapeutics. We purified endogenous RyR2 from porcine heart using glutathione S-transferase–fused FKBP12. To obtain the structure of RyR2 in the closed state, 5 mM EDTA was included throughout purification. To capture an open RyR2, the protein was purified in the presence of 20 μM Ca2+ and the compound 2,2′,3,5′,6-pentachlorobiphenyl (PCB95) that can stabilize RyR1 in the open state. RESULTS The electron microscopy (EM) maps for RyR2 were reconstructed to 4.4- and 4.2-Å resolutions for the closed and open states, respectively. Compared to the structure of RyR1, a number of armadillo repeats in the C terminus of the helical domain 2 were invisible in RyR2, likely due to intrinsic flexibility. At 20 μM Ca2+, the cytoplasmic gate in the closed structure is dilated by approximately 8 Å, resulting in the shift of the constriction site from Ile4868 to Gln4864. The four Gln4864 residues enclose a gate with a diameter of approximately 4 Å, allowing Ca2+ passage in a single file. CONCLUSION Structure comparison of the open and closed RyR2 shows little intradomain rearrangement of the armadillo-containing cytoplasmic domains including the amino terminal domain, the Handle domain, and the Helical domain. Relative shifts between these domains result in the breathing motion of the periphery of the cytoplasmic canopy and the rotation of the Central domain. The horseshoe-shaped Central domain, with its convex side interacting with the three armadillo domains and its concave side wrapping around the cytoplasmic O-ring of the channel domain, serves as the primary transducer that integrates and translates the conformational changes of the cytoplasmic domains to channel gating. However, the mechanism of Ca2+ sensing and activation of RyR2 remains to be elucidated. Cryogenic EM structures of RyR2 from porcine heart in both the closed and open states at near-atomic resolutions. (Top) Representative two-dimensional class averages of electron micrographs of the closed and open RyR2. (Bottom) The two structures are superimposed relative to the transmembrane domain. The blue arrows indicate the overall shifts of the cytoplasmic region from the closed state to the open state. SR, sarcoplasmic reticulum. RyR2 is a high-conductance intracellular calcium (Ca2+) channel that controls the release of Ca2+ from the sarco(endo)plasmic reticulum of a variety of cells. Here, we report the structures of RyR2 from porcine heart in both the open and closed states at near-atomic resolutions determined using single-particle electron cryomicroscopy. Structural comparison reveals a breathing motion of the overall cytoplasmic region resulted from the interdomain movements of amino-terminal domains (NTDs), Helical domains, and Handle domains, whereas almost no intradomain shifts are observed in these armadillo repeats–containing domains. Outward rotations of the Central domains, which integrate the conformational changes of the cytoplasmic region, lead to the dilation of the cytoplasmic gate through coupled motions. Our structural and mutational characterizations provide important insights into the gating and disease mechanism of RyRs.


Journal of Biological Chemistry | 2015

Roles of the NH2-terminal Domains of Cardiac Ryanodine Receptor in Ca2+ Release Activation and Termination

Yingjie Liu; Bo Sun; Zhichao Xiao; Ruiwu Wang; Wenting Guo; Joe Z. Zhang; Tao Mi; Yundi Wang; Peter P. Jones; Filip Van Petegem; S. R. Wayne Chen

Background: The NH2-terminal region of cardiac ryanodine receptor (RyR2) contains three domains (A, B, and C) that harbor many disease-causing mutations. Results: Domains A, B, and C distinctively regulate the activation and termination of Ca2+ release. Conclusion: Individual NH2-terminal domains play distinct roles in RyR2 channel function. Significance: These data shed new insights into the actions of RyR2 NH2-terminal disease mutations. The NH2-terminal region (residues 1–543) of the cardiac ryanodine receptor (RyR2) harbors a large number of mutations associated with cardiac arrhythmias and cardiomyopathies. Functional studies have revealed that the NH2-terminal region is involved in the activation and termination of Ca2+ release. The three-dimensional structure of the NH2-terminal region has recently been solved. It is composed of three domains (A, B, and C). However, the roles of these individual domains in Ca2+ release activation and termination are largely unknown. To understand the functional significance of each of these NH2-terminal domains, we systematically deleted these domains and assessed their impact on caffeine- or Ca2+-induced Ca2+ release and store overload-induced Ca2+ release (SOICR) in HEK293 cells. We found that all deletion mutants were capable of forming caffeine- and ryanodine-sensitive functional channels, indicating that the NH2-terminal region is not essential for channel gating. Ca2+ release measurements revealed that deleting domain A markedly reduced the threshold for SOICR termination but had no effect on caffeine or Ca2+ activation or the threshold for SOICR activation, whereas deleting domain B substantially enhanced caffeine and Ca2+ activation and lowered the threshold for SOICR activation and termination. Conversely, deleting domain C suppressed caffeine activation, abolished Ca2+ activation and SOICR, and diminished protein expression. These results suggest that domain A is involved in channel termination, domain B is involved in channel suppression, and domain C is critical for channel activation and expression. Our data shed new insights into the structure-function relationship of the NH2-terminal domains of RyR2 and the action of NH2-terminal disease mutations.


The Journal of General Physiology | 2017

Control of cardiac ryanodine receptor by sarcoplasmic reticulum luminal Ca2

Peter P. Jones; Wenting Guo; S. R. Wayne Chen

Jones et al. propose that SR luminal Ca2+ regulates RyR2 activity via a luminal Ca2+ sensor distinct from the cytosolic Ca2+ sensor.


Journal of Biological Chemistry | 2016

Enhanced Cytosolic Ca2+ Activation Underlies a Common Defect of Central Domain Cardiac Ryanodine Receptor Mutations Linked to Arrhythmias

Zhichao Xiao; Wenting Guo; Bo Sun; Donald J. Hunt; Jinhong Wei; Yingjie Liu; Yundi Wang; Ruiwu Wang; Peter P. Jones; Thomas G. Back; S. R. Wayne Chen

Recent three-dimensional structural studies reveal that the central domain of ryanodine receptor (RyR) serves as a transducer that converts long-range conformational changes into the gating of the channel pore. Interestingly, the central domain encompasses one of the mutation hotspots (corresponding to amino acid residues 3778–4201) that contains a number of cardiac RyR (RyR2) mutations associated with catecholaminergic polymorphic ventricular tachycardia (CPVT) and atrial fibrillation (AF). However, the functional consequences of these central domain RyR2 mutations are not well understood. To gain insights into the impact of the mutation and the role of the central domain in channel function, we generated and characterized eight disease-associated RyR2 mutations in the central domain. We found that all eight central domain RyR2 mutations enhanced the Ca2+-dependent activation of [3H]ryanodine binding, increased cytosolic Ca2+-induced fractional Ca2+ release, and reduced the activation and termination thresholds for spontaneous Ca2+ release in HEK293 cells. We also showed that racemic carvedilol and the non-beta-blocking carvedilol enantiomer, (R)-carvedilol, suppressed spontaneous Ca2+ oscillations in HEK293 cells expressing the central domain RyR2 mutations associated with CPVT and AF. These data indicate that the central domain is an important determinant of cytosolic Ca2+ activation of RyR2. These results also suggest that altered cytosolic Ca2+ activation of RyR2 represents a common defect of RyR2 mutations associated with CPVT and AF, which could potentially be suppressed by carvedilol or (R)-carvedilol.


PLOS ONE | 2015

The H29D Mutation Does Not Enhance Cytosolic Ca2+ Activation of the Cardiac Ryanodine Receptor

Zhichao Xiao; Wenting Guo; Siobhan M. Wong King Yuen; Ruiwu Wang; Lin Zhang; Filip Van Petegem; S. R. Wayne Chen

The N-terminal domain of the cardiac ryanodine receptor (RyR2) harbors a large number of naturally occurring mutations that are associated with stress-induced ventricular tachyarrhythmia and sudden death. Nearly all these disease-associated N-terminal mutations are located at domain interfaces or buried within domains. Mutations at these locations would alter domain-domain interactions or the stability/folding of domains. Recently, a novel RyR2 mutation H29D associated with ventricular arrhythmia at rest was found to enhance the activation of single RyR2 channels by diastolic levels of cytosolic Ca2+. Unlike other N-terminal disease-associated mutations, the H29D mutation is located on the surface of the N-terminal domain. It is unclear how this surface-exposed H29D mutation that does not appear to interact with other parts of the RyR2 structure could alter the intrinsic properties of the channel. Here we carried out detailed functional characterization of the RyR2-H29D mutant at the molecular and cellular levels. We found that the H29D mutation has no effect on the basal level or the Ca2+ dependent activation of [3H]ryanodine binding to RyR2, the cytosolic Ca2+ activation of single RyR2 channels, or the cytosolic Ca2+- or caffeine-induced Ca2+ release in HEK293 cells. In addition, the H29D mutation does not alter the propensity for spontaneous Ca2+ release or the thresholds for Ca2+ release activation or termination. Furthermore, the H29D mutation does not have significant impact on the thermal stability of the N-terminal region (residues 1–547) of RyR2. Collectively, our data show that the H29D mutation exerts little or no effect on the function of RyR2 or on the folding stability of the N-terminal region. Thus, our results provide no evidence that the H29D mutation enhances the cytosolic Ca2+ activation of RyR2.


Heart Rhythm | 2018

Linking the heart and the brain: Neurodevelopmental disorders in patients with catecholaminergic polymorphic ventricular tachycardia

Krystien V.V. Lieve; Judith M.A. Verhagen; Jinhong Wei; J. Martijn Bos; Christian van der Werf; Ferran Rosés i Noguer; Grazia M.S. Mancini; Wenting Guo; Ruiwu Wang; Freek van den Heuvel; Ingrid M.E. Frohn-Mulder; Wataru Shimizu; Akihiko Nogami; Hitoshi Horigome; Jason D. Roberts; Antoine Leenhardt; Harry J.G.M. Crijns; Andreas C. Blank; Takeshi Aiba; Ans C.P. Wiesfeld; Nico A. Blom; Naokata Sumitomo; Jan Till; Michael J. Ackerman; S. R. Wayne Chen; Ingrid van de Laar; Arthur A.M. Wilde

BACKGROUND Catecholaminergic polymorphic ventricular tachycardia (CPVT) is an uncommon inherited arrhythmia disorder characterized by adrenergically evoked ventricular arrhythmias. Mutations in the cardiac calcium release channel/ryanodine receptor gene (RYR2) are identified in the majority of patients with CPVT. RyR2 is also the major RyR isoform expressed in the brain. OBJECTIVE The purpose of this study was to estimate the prevalence of intellectual disability (ID) and other neurodevelopmental disorders (NDDs) in RYR2-associated CPVT (CPVT1) and to study the characteristics of these patients. METHODS We reviewed the medical records of all CPVT1 patients from 12 international centers and analyzed the characteristics of all CPVT1 patients with concomitant NDDs. We functionally characterized the mutations to assess their response to caffeine activation. We did not correct for potential confounders. RESULTS Among 421 CPVT1 patients, we identified 34 patients with ID (8%; 95% confidence interval 6%-11%). Median age at diagnosis was 9.3 years (interquartile range 7.0-14.5). Parents for 24 of 34 patients were available for genetic testing, and 13 of 24 (54%) had a de novo mutation. Severity of ID ranged from mild to severe and was accompanied by other NDDs in 9 patients (26%). Functionally, the ID-associated mutations showed a markedly enhanced response of RyR2 to activation by caffeine. Seventeen patients (50%) also had supraventricular arrhythmias. During median follow-up of 8.4 years (interquartile range 1.8-12.4), 15 patients (45%) experienced an arrhythmic event despite adequate therapy. CONCLUSION Our study indicates that ID is more prevalent among CPVT1 patients (8%) than in the general population (1%-3%). This subgroup of CPVT1 patients reveals a malignant cardiac phenotype with marked supraventricular and ventricular arrhythmias.


European Journal of Human Genetics | 2018

De novo ITPR1 variants are a recurrent cause of early-onset ataxia, acting via loss of channel function

Matthis Synofzik; Katherine L. Helbig; Florian Harmuth; Tine Deconinck; Pranoot Tanpaiboon; Bo Sun; Wenting Guo; Ruiwu Wang; Erika Palmaer; Sha Tang; G. Bradley Schaefer; Janina Gburek-Augustat; Stephan Züchner; Ingeborg Krägeloh-Mann; Jonathan Baets; Peter Bauer; S. R. Wayne Chen; Ludger Schöls; Rebecca Schüle

We explored the clinico-genetic basis of spinocerebellar ataxia 29 (SCA29) by determining the frequency, phenotype, and functional impact of ITPR1 missense variants associated with early-onset ataxia (EOA). Three hundred thirty one patients from a European EOA target cohort (n = 120), US-American EOA validation cohort (n = 72), and early-onset epileptic encephalopathy (EOEE) control cohort (n = 139) were screened for de novo ITPR1 variants. The target cohort was also screened for inherited ITPR1 variants. The variants’ functional impact was determined by IP3-induced Ca2+ release in HEK293 cells. 3/120 patients (2.5%) from the target cohort and 4/72 patients (5.5%) from the validation cohort, but none from the EOEE control cohort, carried de novo ITPR1 variants. However, most ITPR1 variants (7/10 = 70%) in the target cohort were inherited from a healthy parent, with 3/6 patients carrying disease-causing variants in other genes. This suggests limited or no phenotypic impact of many ITPR1 missense variants, even if ultra-rare and well-conserved. While common bioinformatics tools did not discriminate de novo from other ITPR1 variants, functional characterization demonstrated reduced IP3-induced Ca2+ release for all de novo variants, including the recurrent c.805C>T (p.(R269W)) variant. In sum, these findings show that de novo ITPR1 missense variants are a recurrent cause of EOA (SCA29) across independent cohorts, acting via loss of IP3 channel function. Inherited ITPR1 variants are also enriched in EOA, but often without strong impact, albeit rare and well-conserved. Functional studies allow identifying ITPR1 variants with large impact, likely disease-causing. Such functional confirmation is warranted for inherited ITPR1 variants before making a SCA29 diagnosis.


Biochemical Journal | 2018

Reduced expression of cardiac ryanodine receptor protects against stress-induced ventricular tachyarrhythmia, but increases the susceptibility to cardiac alternans

Xiaowei Zhong; Alexander Vallmitjana; Bo Sun; Zhichao Xiao; Wenting Guo; Jinhong Wei; Mingke Ni; Yongxiang Chen; Edward R. O'Brien; Anne M. Gillis; Masahiko Hoshijima; Hiroshi Takeshima; Leif Hove-Madsen; Raul Benitez; Darrell Belke; S. R. Wayne Chen

Reduced protein expression of the cardiac ryanodine receptor type 2 (RyR2) is thought to affect the susceptibility to stress-induced ventricular tachyarrhythmia (VT) and cardiac alternans, but direct evidence for the role of RyR2 protein expression in VT and cardiac alternans is lacking. Here, we used a mouse model (crrm1) that expresses a reduced level of the RyR2 protein to determine the impact of reduced RyR2 protein expression on the susceptibility to VT, cardiac alternans, cardiac hypertrophy, and sudden death. Electrocardiographic analysis revealed that after the injection of relatively high doses of caffeine and epinephrine (agents commonly used for stress test), wild-type (WT) mice displayed long-lasting VTs, whereas the crrm1 mutant mice exhibited no VTs at all, indicating that the crrm1 mutant mice are resistant to stress-induced VTs. Intact heart Ca2+ imaging and action potential (AP) recordings showed that the crrm1 mutant mice are more susceptible to fast-pacing induced Ca2+ alternans and AP duration alternans compared with WT mice. The crrm1 mutant mice also showed an increased heart-to-body-weight ratio and incidence of sudden death at young ages. Furthermore, the crrm1 mutant hearts displayed altered Ca2+ transients with increased time-to-peak and decay time (T50), increased ventricular wall thickness and ventricular cell area compared with WT hearts. These results indicate that reduced RyR2 protein expression suppresses stress-induced VTs, but enhances the susceptibility to cardiac alternans, hypertrophy, and sudden death.


Biochemical Journal | 2017

Reduced threshold for store overload-induced Ca 2+ release is a common defect of RyR1 mutations associated with malignant hyperthermia and central core disease

Wenqian Chen; Andrea Koop; Yingjie Liu; Wenting Guo; Jinhong Wei; Ruiwu Wang; David H. MacLennan; Robert T. Dirksen; Sui Rong Wayne Chen

Mutations in the skeletal muscle ryanodine receptor (RyR1) cause malignant hyperthermia (MH) and central core disease (CCD), whereas mutations in the cardiac ryanodine receptor (RyR2) lead to catecholaminergic polymorphic ventricular tachycardia (CPVT). Most disease-associated RyR1 and RyR2 mutations are located in the N-terminal, central, and C-terminal regions of the corresponding ryanodine receptor (RyR) isoform. An increasing body of evidence demonstrates that CPVT-associated RyR2 mutations enhance the propensity for spontaneous Ca2+ release during store Ca2+ overload, a process known as store overload-induced Ca2+ release (SOICR). Considering the similar locations of disease-associated RyR1 and RyR2 mutations in the RyR structure, we hypothesize that like CPVT-associated RyR2 mutations, MH/CCD-associated RyR1 mutations also enhance SOICR. To test this hypothesis, we determined the impact on SOICR of 12 MH/CCD-associated RyR1 mutations E2347-del, R2163H, G2434R, R2435L, R2435H, and R2454H located in the central region, and Y4796C, T4826I, L4838V, A4940T, G4943V, and P4973L located in the C-terminal region of the channel. We found that all these RyR1 mutations reduced the threshold for SOICR. Dantrolene, an acute treatment for MH, suppressed SOICR in HEK293 cells expressing the RyR1 mutants R164C, Y523S, R2136H, R2435H, and Y4796C. Interestingly, carvedilol, a commonly used β-blocker that suppresses RyR2-mediated SOICR, also inhibits SOICR in these RyR1 mutant HEK293 cells. Therefore, these results indicate that a reduced SOICR threshold is a common defect of MH/CCD-associated RyR1 mutations, and that carvedilol, like dantrolene, can suppress RyR1-mediated SOICR. Clinical studies of the effectiveness of carvedilol as a long-term treatment for MH/CCD or other RyR1-associated disorders may be warranted.


Biochemical Journal | 2015

Role of Cys³⁶⁰² in the function and regulation of the cardiac ryanodine receptor.

Tao Mi; Zhichao Xiao; Wenting Guo; Yijun Tang; Florian Hiess; Jianmin Xiao; Yundi Wang; Joe Z. Zhang; Lin Zhang; Ruiwu Wang; Peter P. Jones; S. R. Wayne Chen

Collaboration


Dive into the Wenting Guo's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Filip Van Petegem

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Katherine L. Helbig

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar

Sha Tang

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lin Zhang

University of Calgary

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge