Network


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

Hotspot


Dive into the research topics where Zhong Guo is active.

Publication


Featured researches published by Zhong Guo.


Nature Chemical Biology | 2009

Analysis of the eukaryotic prenylome by isoprenoid affinity tagging

Uyen T. T. Nguyen; Zhong Guo; Christine Delon; Yao-Wen Wu; Céline Deraeve; Benjamin Fränzel; Robin S. Bon; Wulf Blankenfeldt; Roger S. Goody; Herbert Waldmann; Dirk Wolters; Kirill Alexandrov

Protein prenylation is a widespread phenomenon in eukaryotic cells that affects many important signaling molecules. We describe the structure-guided design of engineered protein prenyltransferases and their universal synthetic substrate, biotin-geranylpyrophosphate. These new tools allowed us to detect femtomolar amounts of prenylatable proteins in cells and organs and to identify their cognate protein prenyltransferases. Using this approach, we analyzed the in vivo effects of protein prenyltransferase inhibitors. Whereas some of the inhibitors displayed the expected activities, others lacked in vivo activity or targeted a broader spectrum of prenyltransferases than previously believed. To quantitate the in vivo effect of the prenylation inhibitors, we profiled biotin-geranyl-tagged RabGTPases across the proteome by mass spectrometry. We also demonstrate that sites of active vesicular transport carry most of the RabGTPases. This approach enables a quantitative proteome-wide analysis of the regulation of protein prenylation and its modulation by therapeutic agents.


Journal of Biological Chemistry | 2014

Cortactin scaffolds Arp2/3 and WAVE2 at the epithelial zonula adherens.

Siew Ping Han; Yann Gambin; Guillermo A. Gomez; Suzie Verma; Nichole Giles; Magdalene Michael; Selwin K. Wu; Zhong Guo; Wayne A. Johnston; Emma Sierecki; Robert G. Parton; Kirill Alexandrov; Alpha S. Yap

Background: Productive epithelial interactions require actin filament assembly at E-cadherin adhesions. Results: Cortactin localizes to the zonula adherens through interactions with E-cadherin and N-WASP; there it recruits Arp2/3 and WAVE2 by separate mechanisms to support actin nucleation. Conclusion: Cortactin acts as a coincident scaffold. Significance: Cortactin can regulate the dynamic integration of cadherin adhesion with the actin cytoskeleton. Cadherin junctions arise from the integrated action of cell adhesion, signaling, and the cytoskeleton. At the zonula adherens (ZA), a WAVE2-Arp2/3 actin nucleation apparatus is necessary for junctional tension and integrity. But how this is coordinated with cadherin adhesion is not known. We now identify cortactin as a key scaffold for actin regulation at the ZA, which localizes to the ZA through influences from both E-cadherin and N-WASP. Using cell-free protein expression and fluorescent single molecule coincidence assays, we demonstrate that cortactin binds directly to the cadherin cytoplasmic tail. However, its concentration with cadherin at the apical ZA also requires N-WASP. Cortactin is known to bind Arp2/3 directly (Weed, S. A., Karginov, A. V., Schafer, D. A., Weaver, A. M., Kinley, A. W., Cooper, J. A., and Parsons, J. T. (2000) J. Cell Biol. 151, 29–40). We further show that cortactin can directly bind WAVE2, as well as Arp2/3, and both these interactions are necessary for actin assembly at the ZA. We propose that cortactin serves as a platform that integrates regulators of junctional actin assembly at the ZA.


The EMBO Journal | 2008

Structures of RabGGTase-substrate/product complexes provide insights into the evolution of protein prenylation.

Zhong Guo; Yao-Wen Wu; Debapratim Das; Christine Delon; Janinna Cramer; Shen Yu; Sandra Thuns; Nataliya Lupilova; Herbert Waldmann; Luc Brunsveld; Roger S. Goody; Kirill Alexandrov; Wulf Blankenfeldt

Post‐translational isoprenylation of proteins is carried out by three related enzymes: farnesyltransferase, geranylgeranyl transferase‐I, and Rab geranylgeranyl transferase (RabGGTase). Despite the fact that the last one is responsible for the largest number of individual protein prenylation events in the cell, no structural information is available on its interaction with substrates and products. Here, we present structural and biophysical analyses of RabGGTase in complex with phosphoisoprenoids as well as with the prenylated peptides that mimic the C terminus of Rab7 GTPase. The data demonstrate that, unlike other protein prenyl transferases, both RabGGTase and its substrate RabGTPases completely ‘outsource’ their specificity for each other to an accessory subunit, the Rab escort protein (REP). REP mediates the placement of the C terminus of RabGTPase into the active site of RabGGTase through a series protein–protein interactions of decreasing strength and selectivity. This arrangement enables RabGGTase to prenylate any cysteine‐containing sequence. On the basis of our structural and thermodynamic data, we propose that RabGGTase has evolved from a GGTase‐I‐like molecule that ‘learned’ to interact with a recycling factor (GDI) that, in turn, eventually gave rise to REP.


Journal of Biological Chemistry | 2012

Quantitative Analysis of Prenylated RhoA Interaction with Its Chaperone, RhoGDI.

Zakir Tnimov; Zhong Guo; Yann Gambin; Uyen T. T. Nguyen; Yao-Wen Wu; Daniel Abankwa; Anouk Stigter; Brett M. Collins; Herbert Waldmann; Roger S. Goody; Kirill Alexandrov

Background: RhoGDI is a key regulator and a chaperon of Rho GTPases. Results: RhoGDI strongly discriminates between GDP- and GTP-bound forms of prenylated RhoA, although both complexes are of high affinity. Conclusion: We provide direct evidence for the existence of two populations of the RhoGDI·RhoA complexes in the cell, characterized by different lifetimes. Significance: The obtained data allows us to formulate the model for membrane delivery and extraction of Rho GTPases. Small GTPases of the Rho family regulate cytoskeleton remodeling, cell polarity, and transcription, as well as the cell cycle, in eukaryotic cells. Membrane delivery and recycling of the Rho GTPases is mediated by Rho GDP dissociation inhibitor (RhoGDI), which forms a stable complex with prenylated Rho GTPases. We analyzed the interaction of RhoGDI with the active and inactive forms of prenylated and unprenylated RhoA. We demonstrate that RhoGDI binds the prenylated form of RhoA·GDP with unexpectedly high affinity (Kd = 5 pm). The very long half-life of the complex is reduced 25-fold on RhoA activation, with a concomitant reduction in affinity (Kd = 3 nm). The 2.8-Å structure of the RhoA·guanosine 5′-[β,γ-imido] triphosphate (GMPPNP)·RhoGDI complex demonstrated that complex formation forces the activated RhoA into a GDP-bound conformation in the absence of nucleotide hydrolysis. We demonstrate that membrane extraction of Rho GTPase by RhoGDI is a thermodynamically favored passive process that operates through a series of progressively tighter intermediates, much like the one that is mediated by RabGDI.


Journal of the American Chemical Society | 2012

Psoromic Acid is a Selective and Covalent Rab-Prenylation Inhibitor Targeting Autoinhibited RabGGTase

Céline Deraeve; Zhong Guo; Robin S. Bon; Wulf Blankenfeldt; Raffaella DiLucrezia; Alexander Wolf; Sascha Menninger; E. Anouk Stigter; Stefan Wetzel; Axel Choidas; Kirill Alexandrov; Herbert Waldmann; Roger S. Goody; Yao-Wen Wu

Post-translational attachment of geranylgeranyl isoprenoids to Rab GTPases, the key organizers of intracellular vesicular transport, is essential for their function. Rab geranylgeranyl transferase (RabGGTase) is responsible for prenylation of Rab proteins. Recently, RabGGTase inhibitors have been proposed to be potential therapeutics for treatment of cancer and osteoporosis. However, the development of RabGGTase selective inhibitors is complicated by its structural and functional similarity to other protein prenyltransferases. Herein we report identification of the natural product psoromic acid (PA) that potently and selectively inhibits RabGGTase with an IC(50) of 1.3 μM. Structure-activity relationship analysis suggested a minimal structure involving the depsidone core with a 3-hydroxyl and 4-aldehyde motif for binding to RabGGTase. Analysis of the crystal structure of the RabGGTase:PA complex revealed that PA forms largely hydrophobic interactions with the isoprenoid binding site of RabGGTase and that it attaches covalently to the N-terminus of the α subunit. We found that in contrast to other protein prenyltransferases, RabGGTase is autoinhibited through N-terminal (α)His2 coordination with the catalytic zinc ion. Mutation of (α)His dramatically enhances the reaction rate, indicating that the activity of RabGGTase is likely regulated in vivo. The covalent binding of PA to the N-terminus of the RabGGTase α subunit seems to potentiate its interaction with the active site and explains the selectivity of PA for RabGGTase. Therefore, psoromic acid provides a new starting point for the development of selective RabGGTase inhibitors.


Journal of Biological Chemistry | 2013

Intermediates in the guanine nucleotide exchange reaction of Rab8 protein catalyzed by guanine nucleotide exchange factors Rabin8 and GRAB.

Zhong Guo; Xiaomin Hou; Roger S. Goody; Aymelt Itzen

Background: The GEFs Rabin8 and GRAB are activators of the vesicular trafficking regulator Rab8. Results: The catalytic mechanism of Rabin8/GRAB in Rab8 has been elucidated in biophysical and structural detail. Conclusion: Rabin8 and GRAB are catalytically moderately efficient enzymes and act by disturbing Mg2+ binding and Rab8-guanine base interactions. Significance: Obtaining snapshots of the nucleotide exchange reaction is crucial to understanding the mechanism of Rab GEFs. Small G-proteins of the Ras superfamily control the temporal and spatial coordination of intracellular signaling networks by acting as molecular on/off switches. Guanine nucleotide exchange factors (GEFs) regulate the activation of these G-proteins through catalytic replacement of GDP by GTP. During nucleotide exchange, three distinct substrate·enzyme complexes occur: a ternary complex with GDP at the start of the reaction (G-protein·GEF·GDP), an intermediary nucleotide-free binary complex (G-protein·GEF), and a ternary GTP complex after productive G-protein activation (G-protein·GEF·GTP). Here, we show structural snapshots of the full nucleotide exchange reaction sequence together with the G-protein substrates and products using Rabin8/GRAB (GEF) and Rab8 (G-protein) as a model system. Together with a thorough enzymatic characterization, our data provide a detailed view into the mechanism of Rabin8/GRAB-mediated nucleotide exchange.


Journal of Medicinal Chemistry | 2012

Development of Selective, Potent RabGGTase Inhibitors

Anouk Stigter; Zhong Guo; Robin S. Bon; Yao-Wen Wu; Axel Choidas; Alexander Wolf; Sascha Menninger; Herbert Waldmann; Wulf Blankenfeldt; Roger S. Goody

Members of the Ras superfamily of small GTPases are frequently mutated in cancer. Therefore, inhibitors have been developed to address the acitivity of these GTPases by inhibiting their prenylating enzymes FTase, GGTase I, and RabGGTase. In contrast to FTase and GGTase I, only a handful of RabGGTase inhibitors have been developed. The most active RabGGTase inhibitor known until recently was an FTase inhibitor which hit RabGGTase as an off-target. We recently reported our efforts to tune the selectivity of these inhibitors toward RabGGTase. Here we describe an extended set of selective inhibitors. The requirements for selective RabGGTase inhibitors are described in detail, guided by multiple crystal structures. In order to relate in vitro and cellular activity, a high-throughput assay system to detect the attachment of [(3)H]geranylgeranyl groups to Rab was used. Selective RabGGTase inhibition allows the establishment of novel drug discovery programs aimed at the development of anticancer therapeutics.


Angewandte Chemie | 2011

Structure‐Guided Development of Selective RabGGTase Inhibitors

Robin S. Bon; Zhong Guo; E. Anouk Stigter; Stefan Wetzel; Sascha Menninger; Alexander Wolf; Axel Choidas; Kirill Alexandrov; Wulf Blankenfeldt; Roger S. Goody; Herbert Waldmann

Designing for selectivity: A combination of protein crystal‐structure analysis, virtual screening, and synthetic chemistry has been used to develop noncytotoxic inhibitors of RabGGTase (IC50: 42 nM for the example shown; red O, blue N, yellow S) that are selective over FTase and GGTase I. Furthermore, the inhibitors display cellular activity and inhibit cancer cell proliferation.


Journal of Medicinal Chemistry | 2009

Design, Synthesis, and Characterization of Peptide-Based Rab Geranylgeranyl Transferase Inhibitors

Kui-Thong Tan; Robin S. Bon; Zhong Guo; Christine Delon; Stefan Wetzel; Sabine Arndt; Kirill Alexandrov; Herbert Waldmann; Roger S. Goody; Yao-Wen Wu; Wulf Blankenfeldt

Rab geranylgeranyl transferase (RabGGTase) catalyzes the attachment of geranylgeranyl isoprenoids to Rab guanine triphosphatases, which are key regulators in vesicular transport. Because geranylgeranylation is required for proper function and overexpression of Rabs has been observed in various cancers, RabGGTase may be a target for novel therapeutics. The development of selective inhibitors is, however, difficult because two related enzymes involved in other cellular processes exist in eukaryotes and because RabGGTase recognizes protein substrates indirectly, resulting in relaxed specificity. We report the synthesis of a peptidic library based on the farnesyl transferase inhibitor pepticinnamin E. Of 469 compounds investigated, several were identified as selective for RabGGTase with low micromolar IC(50) values. The compounds were not generally cytotoxic and inhibited Rab isoprenylation in COS-7 cells. Crystal structure analysis revealed that selective inhibitors interact with a tunnel unique to RabGGTase, implying that this structural motif is an attractive target for improved RabGGTase inhibitors.


Angewandte Chemie | 2008

Development of selective RabGGTase inhibitors and crystal structure of a RabGGTase-inhibitor complex.

Zhong Guo; Yao-Wen Wu; Kui-Thong Tan; Robin S. Bon; Christine Delon; Uyen T. T. Nguyen; Stefan Wetzel; Sabine Arndt; Roger S. Goody; Wulf Blankenfeldt; Kirill Alexandrov; Herbert Waldmann

Stopping the transfer: Based on the structure of pepticinnamin E, specific inhibitors of Rab geranylgeranyl transferase (RabGGTase) with activity in cells were developed, and the first crystal structure of the enzyme in complex with an inhibitor is reported (see inhibitor structure and positioning in the active site of the enzyme). The findings may have implications for the chemical‐biological study of Rab prenylation and vesicular transport and the involvement of RabGGTase in the establishment of disease.

Collaboration


Dive into the Zhong Guo's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Wulf Blankenfeldt

Braunschweig University of Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lin Luo

University of Queensland

View shared research outputs
Researchain Logo
Decentralizing Knowledge