Network


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

Hotspot


Dive into the research topics where Guolin Ma is active.

Publication


Featured researches published by Guolin Ma.


Nature Cell Biology | 2015

Proteomic mapping of ER-PM junctions identifies STIMATE as a regulator of Ca2+ influx

Ji Jing; Lian He; Aomin Sun; Ariel Quintana; Yue-He Ding; Guolin Ma; Peng Tan; Xiaowen Liang; Xiaolu Zheng; Liangyi Chen; Xiaodong Shi; Shenyuan L. Zhang; Ling Zhong; Yun Huang; Meng-Qiu Dong; Cheryl L. Walker; Patrick G. Hogan; Youjun Wang; Yubin Zhou

Specialized junctional sites that connect the plasma membrane (PM) and endoplasmic reticulum (ER) play critical roles in controlling lipid metabolism and Ca2+ signalling. Store-operated Ca2+ entry mediated by dynamic STIM1–ORAI1 coupling represents a classical molecular event occurring at ER–PM junctions, but the protein composition and how previously unrecognized protein regulators facilitate this process remain ill-defined. Using a combination of spatially restricted biotin labelling in situ coupled with mass spectrometry and a secondary screen based on bimolecular fluorescence complementation, we mapped the proteome of intact ER–PM junctions in living cells without disrupting their architectural integrity. Our approaches led to the discovery of an ER-resident multi-transmembrane protein that we call STIMATE (STIM-activating enhancer, encoded by TMEM110) as a positive regulator of Ca2+ influx in vertebrates. STIMATE physically interacts with STIM1 to promote STIM1 conformational switch. Genetic depletion of STIMATE substantially reduces STIM1 puncta formation at ER–PM junctions and suppresses the Ca2+–NFAT signalling. Our findings enable further genetic studies to elucidate the function of STIMATE in normal physiology and disease, and set the stage to uncover more uncharted functions of hitherto underexplored ER–PM junctions.


Nature Communications | 2015

Inside-out Ca2+ signalling prompted by STIM1 conformational switch

Guolin Ma; Ming Wei; Lian He; Chongxu Liu; Bo Wu; Shenyuan L. Zhang; Ji Jing; Xiaowen Liang; Alessandro Senes; Peng Tan; Siwei Li; Aomin Sun; Yunchen Bi; Ling Zhong; Hongjiang Si; Yuequan Shen; Minyong Li; Mi Sun Lee; Weibin Zhou; Junfeng Wang; Youjun Wang; Yubin Zhou

Store-operated Ca2+ entry mediated by STIM1 and ORAI1 constitutes one of the major Ca2+ entry routes in mammalian cells. The molecular choreography of STIM1–ORAI1 coupling is initiated by endoplasmic reticulum (ER) Ca2+ store depletion with subsequent oligomerization of the STIM1 ER-luminal domain, followed by its redistribution towards the plasma membrane to gate ORAI1 channels. The mechanistic underpinnings of this inside-out Ca2+ signalling were largely undefined. By taking advantage of a unique gain-of-function mutation within the STIM1 transmembrane domain (STIM1-TM), here we show that local rearrangement, rather than alteration in the oligomeric state of STIM1-TM, prompts conformational changes in the cytosolic juxtamembrane coiled-coil region. Importantly, we further identify critical residues within the cytoplasmic domain of STIM1 (STIM1-CT) that entail autoinhibition. On the basis of these findings, we propose a model in which STIM1-TM reorganization switches STIM1-CT into an extended conformation, thereby projecting the ORAI-activating domain to gate ORAI1 channels.


Journal of the American Chemical Society | 2017

Engineered Split-TET2 Enzyme for Inducible Epigenetic Remodeling

Minjung Lee; Jia Li; Yi Liang; Guolin Ma; Jixiang Zhang; Lian He; Yuliang Liu; Qian Li; Minyong Li; Deqiang Sun; Yubin Zhou; Yun Huang

The Ten-eleven translocation (TET) family of 5-methylcytosine (5mC) dioxygenases catalyze the conversion of 5mC into 5-hydroxymethylcytosine (5hmC) and further oxidized species to promote active DNA demethylation. Here we engineered a split-TET2 enzyme to enable temporal control of 5mC oxidation and subsequent remodeling of epigenetic states in mammalian cells. We further demonstrate the use of this chemically inducible system to dissect the correlation between DNA hydroxymethylation and chromatin accessibility in the mammalian genome. This chemical-inducible epigenome remodeling tool will find broad use in interrogating cellular systems without altering the genetic code, as well as in probing the epigenotype-phenotype relations in various biological systems.


Pflügers Archiv: European Journal of Physiology | 2016

Molecular mechanisms underlying inhibition of STIM1-Orai1-mediated Ca 2+ entry induced by 2-aminoethoxydiphenyl borate

Ming Wei; Aomin Sun; Guolin Ma; Lian He; Lijuan Zhou; Shuce Zhang; Jin Liu; Shenyuan L. Zhang; Donald L. Gill; Youjun Wang

Store-operated Ca2+ entry (SOCE) mediated by STIM1 and Orai1 is crucial for Ca2+ signaling and homeostasis in most cell types. 2-Aminoethoxydiphenyl borate (2-APB) is a well-described SOCE inhibitor, but its mechanisms of action remain largely elusive. Here, we show that 2-APB does not affect the dimeric state of STIM1, but enhances the intramolecular coupling between the coiled-coil 1 (CC1) and STIM-Orai-activating region (SOAR) of STIM1, with subsequent reduction in the formation of STIM1 puncta in the absence of Orai1 overexpression. 2-APB also inhibits Orai1 channels, directly inhibiting Ca2+ entry through the constitutively active, STIM1-independent Orai1 mutants, Orai1-P245T and Orai1-V102A. When unbound from STIM1, the constitutively active Orai1-V102C mutant is not inhibited by 2-APB. Thus, we used Orai1-V012C as a tool to examine whether 2-APB can also inhibit the coupling between STIM1 and Orai1. We reveal that the functional coupling between STIM1 and Orai1-V102C is inhibited by 2-APB. This inhibition on coupling is indirect, arising from 2-APB’s action on STIM1, and it is most likely mediated by functional channel residues in the Orai1 N-terminus. Overall, our findings on this two-site inhibition mediated by 2-APB provide new understanding on Orai1-activation by STIM1, important to future drug design.


Angewandte Chemie | 2018

Optogenetic Control of Voltage-Gated Calcium Channels

Guolin Ma; Jindou Liu; Yuepeng Ke; Xin Liu; Minyong Li; Fen Wang; Gang Han; Yun Huang; Youjun Wang; Yubin Zhou

Abstract Voltage‐gated Ca2+ (CaV) channels mediate Ca2+ entry into excitable cells to regulate a myriad of cellular events following membrane depolarization. We report the engineering of RGK GTPases, a class of genetically encoded CaV channel modulators, to enable photo‐tunable modulation of CaV channel activity in excitable mammalian cells. This optogenetic tool (designated optoRGK) tailored for CaV channels could find broad applications in interrogating a wide range of CaV‐mediated physiological processes.


Archive | 2018

Engineered Cross-Linking to Study the Pore Architecture of the CRAC Channel

Guolin Ma; Lian He; Ji Jing; Peng Tan; Yun Huang; Yubin Zhou

ORAI1 constitutes the pore-forming subunit of the calcium release-activated calcium (CRAC) channel, a prototypical store-operated channel that is essential for the activation of cells of the immune system. Here we describe a convenient yet powerful cross-linking approach to examine the pore architecture of CRAC channels using ORAI1 proteins engineered to contain one or two cysteine residues. The generalizable cross-linking in situ approach can also be readily extended to study other integral membrane proteins expressed in various types of cells.


Nature Communications | 2018

Calcium sensing by the STIM1 ER-luminal domain

Aparna Gudlur; Ana Eliza Zeraik; Nupura Hirve; V. Rajanikanth; Andrey A. Bobkov; Guolin Ma; Sisi Zheng; Youjun Wang; Yubin Zhou; Elizabeth A. Komives; Patrick G. Hogan

Stromal interaction molecule 1 (STIM1) monitors ER-luminal Ca2+ levels to maintain cellular Ca2+ balance and to support Ca2+ signalling. The prevailing view has been that STIM1 senses reduced ER Ca2+ through dissociation of bound Ca2+ from a single EF-hand site, which triggers a dramatic loss of secondary structure and dimerization of the STIM1 luminal domain. Here we find that the STIM1 luminal domain has 5–6 Ca2+-binding sites, that binding at these sites is energetically coupled to binding at the EF-hand site, and that Ca2+ dissociation controls a switch to a second structured conformation of the luminal domain rather than protein unfolding. Importantly, the other luminal-domain Ca2+-binding sites interact with the EF-hand site to control physiological activation of STIM1 in cells. These findings fundamentally revise our understanding of physiological Ca2+ sensing by STIM1, and highlight molecular mechanisms that govern the Ca2+ threshold for activation and the steep Ca2+ concentration dependence.Stromal interaction molecule 1 (STIM1) monitors ER-luminal Ca2+ levels to maintain cellular Ca2+ balance. Here the authors find that the STIM1 luminal domain monomer has multiple Ca2+ - binding sites which set the threshold for physiological activation of STIM1 in cells.


Cell Calcium | 2018

CRAC channel-based optogenetics

Nhung T. Nguyen; Guolin Ma; Eena Lin; Brendan D’Souza; Ji Jing; Lian He; Yun Huang; Yubin Zhou

Store-operated Ca²+ entry (SOCE) constitutes a major Ca2+ influx pathway in mammals to regulate a myriad of physiological processes, including muscle contraction, synaptic transmission, gene expression, and metabolism. In non-excitable cells, the Ca²+ release-activated Ca²+ (CRAC) channel, composed of ORAI and stromal interaction molecules (STIM), constitutes a prototypical example of SOCE to mediate Ca2+ entry at specialized membrane contact sites (MCSs) between the endoplasmic reticulum (ER) and the plasma membrane (PM). The key steps of SOCE activation include the oligomerization of the luminal domain of the ER-resident Ca2+ sensor STIM1 upon Ca²+ store depletion, subsequent signal propagation toward the cytoplasmic domain to trigger a conformational switch and overcome the intramolecular autoinhibition, and ultimate exposure of the minimal ORAI-activating domain to directly engage and gate ORAI channels in the plasma membrane. This exquisitely coordinated cellular event is also facilitated by the C-terminal polybasic domain of STIM1, which physically associates with negatively charged phosphoinositides embedded in the inner leaflet of the PM to enable efficient translocation of STIM1 into ER-PM MCSs. Here, we present recent progress in recapitulating STIM1-mediated SOCE activation by engineering CRAC channels with optogenetic approaches. These STIM1-based optogenetic tools make it possible to not only mechanistically recapture the key molecular steps of SOCE activation, but also remotely and reversibly control Ca²+-dependent cellular processes, inter-organellar tethering at MCSs, and transcriptional reprogramming when combined with CRISPR/Cas9-based genome-editing tools.


Archive | 2017

The STIM-Orai Pathway: Light-Operated Ca 2+ Entry Through Engineered CRAC Channels

Guolin Ma; Shufan Wen; Yun Huang; Yubin Zhou

Ca2+ signals regulate a plethora of cellular functions that include muscle contraction, heart beating, hormone secretion, lymphocyte activation, gene expression, and metabolism. To study the impact of Ca2+ signals on biological processes, pharmacological tools and caged compounds have been commonly applied to induce fluctuations of intracellular Ca2+ concentrations. These conventional approaches, nonetheless, lack rapid reversibility and high spatiotemporal resolution. To overcome these disadvantages, we and others have devised a series of photoactivatable genetically encoded Ca2+ actuators (GECAs) by installing light sensitivities into a bona fide highly selective Ca2+ channel, the Ca2+ release-activated Ca2+ (CRAC) channel. Store-operated CRAC channel serves as a major route for Ca2+ entry in many cell types. These GECAs enable remote and precise manipulation of Ca2+ signaling in both excitable and non-excitable cells. When combined with nanotechnology, it becomes feasible to wirelessly photo-modulate Ca2+-dependent activities in vivo. In this chapter, we briefly review most recent advances in engineering CRAC channels to achieve optical control over Ca2+ signaling, outline their design principles and kinetic features, and present exemplary applications of GECAs engineered from CRAC channels.


eLife | 2015

Near-infrared photoactivatable control of Ca2+ signaling and optogenetic immunomodulation

Lian He; Yuanwei Zhang; Guolin Ma; Peng Tan; Zhanjun Li; Shengbing Zang; Xiang Wu; Ji Jing; Shaohai Fang; Lijuan Zhou; Youjun Wang; Yun Huang; Patrick G. Hogan; Gang Han; Yubin Zhou

Collaboration


Dive into the Guolin Ma's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Youjun Wang

Beijing Normal University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Patrick G. Hogan

La Jolla Institute for Allergy and Immunology

View shared research outputs
Top Co-Authors

Avatar

Gang Han

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge