Network


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

Hotspot


Dive into the research topics where Qi Qiao is active.

Publication


Featured researches published by Qi Qiao.


Molecular Cell | 2013

Structural Architecture of the CARMA1/Bcl10/MALT1 Signalosome: Nucleation-Induced Filamentous Assembly

Qi Qiao; Chenghua Yang; Chao Zheng; Lorena Fontan; Liron David; Xiong Yu; Clay Bracken; Monica Rosen; Ari Melnick; Edward H. Egelman; Hao Wu

The CARMA1/Bcl10/MALT1 (CBM) signalosome mediates antigen receptor-induced NF-κB signaling to regulate multiple lymphocyte functions. While CARMA1 and Bcl10 contain caspase recruitment domains (CARDs), MALT1 is a paracaspase with structural similarity to caspases. Here we show that the reconstituted CBM signalosome is a helical filamentous assembly in which substoichiometric CARMA1 nucleates Bcl10 filaments. Bcl10 filament formation is a highly cooperative process whose threshold is sensitized by oligomerized CARMA1 upon receptor activation. In cells, both cotransfected CARMA1/Bcl10 complex and the endogenous CBM signalosome are filamentous morphologically. Combining crystallography, nuclear magnetic resonance, and electron microscopy, we reveal the structure of the Bcl10 CARD filament and the mode of interaction between CARMA1 and Bcl10. Structure-guided mutagenesis confirmed the observed interfaces in Bcl10 filament assembly and MALT1 activation in vitro and NF-κB activation in cells. These data support a paradigm of nucleation-induced signal transduction with threshold response due to cooperativity and signal amplification by polymerization.


The Journal of Allergy and Clinical Immunology | 2017

Ruxolitinib reverses dysregulated T helper cell responses and controls autoimmunity caused by a novel signal transducer and activator of transcription 1 (STAT1) gain-of-function mutation

Katja G. Weinacht; Louis-Marie Charbonnier; Fayhan Alroqi; Ashley S. Plant; Qi Qiao; Hao Wu; Clement Ma; Troy R. Torgerson; Sergio D. Rosenzweig; Thomas A. Fleisher; Luigi D. Notarangelo; Imelda C. Hanson; Lisa R. Forbes; Talal A. Chatila

Background: Gain‐of‐function (GOF) mutations in the human signal transducer and activator of transcription 1 (STAT1) manifest in immunodeficiency and autoimmunity with impaired TH17 cell differentiation and exaggerated responsiveness to type I and II interferons. Allogeneic bone marrow transplantation has been attempted in severely affected patients, but outcomes have been poor. Objective: We sought to define the effect of increased STAT1 activity on T helper cell polarization and to investigate the therapeutic potential of ruxolitinib in treating autoimmunity secondary to STAT1 GOF mutations. Methods: We used in vitro polarization assays, as well as phenotypic and functional analysis of STAT1‐mutated patient cells. Results: We report a child with a novel mutation in the linker domain of STAT1 who had life‐threatening autoimmune cytopenias and chronic mucocutaneous candidiasis. Naive lymphocytes from the affected patient displayed increased TH1 and follicular T helper cell and suppressed TH17 cell responses. The mutation augmented cytokine‐induced STAT1 phosphorylation without affecting dephosphorylation kinetics. Treatment with the Janus kinase 1/2 inhibitor ruxolitinib reduced hyperresponsiveness to type I and II interferons, normalized TH1 and follicular T helper cell responses, improved TH17 differentiation, cured mucocutaneous candidiasis, and maintained remission of immune‐mediated cytopenias. Conclusions: Autoimmunity and infection caused by STAT1 GOF mutations are the result of dysregulated T helper cell responses. Janus kinase inhibitor therapy could represent an effective targeted treatment for long‐term disease control in severely affected patients for whom hematopoietic stem cell transplantation is not available.


Journal of Clinical Investigation | 2016

A DOCK8-WIP-WASp complex links T cell receptors to the actin cytoskeleton

Erin Janssen; Mira Tohmé; Mona Hedayat; Marion Leick; Sudha Kumari; Narayanaswamy Ramesh; Michel J. Massaad; Sumana Ullas; Veronica Azcutia; Christopher C. Goodnow; Katrina L. Randall; Qi Qiao; Hao Wu; Waleed Al-Herz; Dianne Cox; John H. Hartwig; Darrell J. Irvine; Francis W. Luscinskas; Raif S. Geha

Wiskott-Aldrich syndrome (WAS) is associated with mutations in the WAS protein (WASp), which plays a critical role in the initiation of T cell receptor-driven (TCR-driven) actin polymerization. The clinical phenotype of WAS includes susceptibility to infection, allergy, autoimmunity, and malignancy and overlaps with the symptoms of dedicator of cytokinesis 8 (DOCK8) deficiency, suggesting that the 2 syndromes share common pathogenic mechanisms. Here, we demonstrated that the WASp-interacting protein (WIP) bridges DOCK8 to WASp and actin in T cells. We determined that the guanine nucleotide exchange factor activity of DOCK8 is essential for the integrity of the subcortical actin cytoskeleton as well as for TCR-driven WASp activation, F-actin assembly, immune synapse formation, actin foci formation, mechanotransduction, T cell transendothelial migration, and homing to lymph nodes, all of which also depend on WASp. These results indicate that DOCK8 and WASp are in the same signaling pathway that links TCRs to the actin cytoskeleton in TCR-driven actin assembly. Further, they provide an explanation for similarities in the clinical phenotypes of WAS and DOCK8 deficiency.


Molecular Cell | 2017

AID Recognizes Structured DNA for Class Switch Recombination

Qi Qiao; Li Wang; Fei-Long Meng; Joyce K. Hwang; Frederick W. Alt; Hao Wu

Activation-induced cytidine deaminase (AID) initiates both class switch recombination (CSR) and somatic hypermutation (SHM) in antibody diversification. Mechanisms of AID targeting and catalysis remain elusive despite its critical immunological roles andxa0off-target effects in tumorigenesis. Here, we produced active human AID and revealed its preferred recognition and deamination of structured substrates. G-quadruplex (G4)-containing substrates mimicking the mammalian immunoglobulin switch regions are particularly good AID substrates inxa0vitro. By solving crystal structures of maltose binding protein (MBP)-fused AID alone and in complex with deoxycytidine monophosphate, we surprisingly identify a bifurcated substrate-binding surface that explains structured substrate recognition by capturing two adjacent single-stranded overhangs simultaneously. Moreover, G4 substrates induce cooperative AID oligomerization. Structure-based mutations that disrupt bifurcated substrate recognition or oligomerization both compromise CSR in splenic B cells. Collectively, our data implicate intrinsic preference of AID for structured substrates and uncover the importance of G4 recognition and oligomerization of AID in CSR.


Cytokine & Growth Factor Reviews | 2014

The CBM signalosome: Potential therapeutic target for aggressive lymphoma?

Chenghua Yang; Liron David; Qi Qiao; Ermelinda Damko; Hao Wu

The CBM signalosome plays a pivotal role in mediating antigen-receptor induced NF-κB signaling to regulate lymphocyte functions. The CBM complex forms filamentous structure and recruits downstream signaling components to activate NF-κB. MALT1, the protease component in the CBM complex, cleaves key proteins in the feedback loop of the NF-κB signaling pathway and enhances NF-κB activation. The aberrant activity of the CBM complex has been linked to aggressive lymphoma. Recent years have witnessed dramatic progresses in understanding the assembly mechanism of the CBM complex, and advances in the development of targeted therapy for aggressive lymphoma. Here, we will highlight these progresses and give an outlook on the potential translation of this knowledge from bench to bedside for aggressive lymphoma patients.


Proceedings of the National Academy of Sciences of the United States of America | 2017

CIDE domains form functionally important higher-order assemblies for DNA fragmentation

Jae Young Choi; Qi Qiao; Se-Hoon Hong; Chang Min Kim; Jae-Hee Jeong; Yeon-Gil Kim; Yong-Keun Jung; Hao Wu; Hyun Ho Park

Significance Cell death-inducing DFF45-like effector (CIDE) domains, initially identified in apoptotic nucleases, form a highly conserved family with diverse functions ranging from cell death to lipid homeostasis and synaptic regulation. Through structural determination of two CIDE family proteins, Drep2 and Drep4, we found that CIDE domains can form helical oligomers. Our results reveal that such higher-order structures not only are conserved in the CIDE family, but also are critically important for both DNA fragmentation and lipid droplet fusion. Therefore, our findings identify the CIDE domain as a scaffolding component for higher-order structure assembly. Our results expand the importance of higher-order structures from the established field of immune signaling to broader biological functions. Cell death-inducing DFF45-like effector (CIDE) domains, initially identified in apoptotic nucleases, form a family with diverse functions ranging from cell death to lipid homeostasis. Here we show that the CIDE domains of Drosophila and human apoptotic nucleases Drep2, Drep4, and DFF40 all form head-to-tail helical filaments. Opposing positively and negatively charged interfaces mediate the helical structures, and mutations on these surfaces abolish nuclease activation for apoptotic DNA fragmentation. Conserved filamentous structures are observed in CIDE family members involved in lipid homeostasis, and mutations on the charged interfaces compromise lipid droplet fusion, suggesting that CIDE domains represent a scaffold for higher-order assembly in DNA fragmentation and other biological processes such as lipid homeostasis.


Cell Reports | 2018

Mind bomb regulates cell death during TNF signaling by suppressing RIPK1’s cytotoxic potential

Rebecca Feltham; Kunzah Jamal; Tencho Tenev; Gianmaria Liccardi; Isabel Jaco; Celia Monteiro Domingues; Otto Morris; Sidonie Wicky John; Alessandro Annibaldi; Marcella Widya; Conor J. Kearney; Danielle M. Clancy; Paul R. Elliott; Timo Glatter; Qi Qiao; Andrew J. Thompson; Alexey I. Nesvizhskii; Alexander Schmidt; David Komander; Hao Wu; Seamus J. Martin; Pascal Meier

Summary Tumor necrosis factor (TNF) is an inflammatory cytokine that can signal cell survival or cell death. The mechanisms that switch between these distinct outcomes remain poorly defined. Here, we show that the E3 ubiquitin ligase Mind Bomb-2 (MIB2) regulates TNF-induced cell death by inactivating RIPK1 via inhibitory ubiquitylation. Although depletion of MIB2 has little effect on NF-κB activation, it sensitizes cells to RIPK1- and caspase-8-dependent cell death. We find that MIB2 represses the cytotoxic potential of RIPK1 by ubiquitylating lysine residues in the C-terminal portion of RIPK1. Our data suggest that ubiquitin conjugation of RIPK1 interferes with RIPK1 oligomerization and RIPK1-FADD association. Disruption of MIB2-mediated ubiquitylation, either by mutation of MIB2’s E3 activity or RIPK1’s ubiquitin-acceptor lysines, sensitizes cells to RIPK1-mediated cell death. Together, our findings demonstrate that Mind Bomb E3 ubiquitin ligases can function as additional checkpoint of cytokine-induced cell death, selectively protecting cells from the cytotoxic effects of TNF.


Structure | 2017

Understanding CARD Tricks in Apoptosomes

Li Wang; Qi Qiao; Hao Wu

While earlier studies of Apaf-1 holo-apoptosome architecture revealed the spectacular heptameric wheel-like structure formed by Apaf-1, the central CARD disk responsible for caspase-9 recruitment remained incompletely resolved. In a recent issue of Structure, Su etxa0al. (2017) describe a crystal structure of the complex between Apaf-1 CARD and caspase-9 CARD. Together with two recent cryo-EM structures, this work brings us closer to a full view of the holo-apoptosome.


Proceedings of the National Academy of Sciences of the United States of America | 2017

Crystal structure of human IRAK1

Li Wang; Qi Qiao; Ryan Ferrao; Chen Shen; John M. Hatcher; Sara J. Buhrlage; Nathanael S. Gray; Hao Wu

Significance Innate immune signaling has an essential role in inflammation, and dysfunction of signaling components in these pathways contributes to autoimmunity and cancer. Interleukin-1 receptor-associated kinase (IRAK) family members are key mediators of signal transduction by Toll-like receptors and Interleukin-1 receptors in innate immunity and therefore serve as potential therapeutic targets for these diseases. The crystal structure of the IRAK1 kinase domain in complex with a small molecule inhibitor reveals important structural details of the kinase that provide insights into the design of selective IRAK inhibitors. Characterization of IRAK1 heterodimerization with the upstream kinase IRAK4 suggests a mechanism of IRAK1 activation by IRAK4. Interleukin 1 (IL-1) receptor-associated kinases (IRAKs) are serine/threonine kinases that play critical roles in initiating innate immune responses against foreign pathogens and other types of dangers through their role in Toll-like receptor (TLR) and interleukin 1 receptor (IL-1R) mediated signaling pathways. Upon ligand binding, TLRs and IL-1Rs recruit adaptor proteins, such as myeloid differentiation primary response gene 88 (MyD88), to the membrane, which in turn recruit IRAKs via the death domains in these proteins to form the Myddosome complex, leading to IRAK kinase activation. Despite their biological and clinical significance, only the IRAK4 kinase domain structure has been determined among the four IRAK family members. Here, we report the crystal structure of the human IRAK1 kinase domain in complex with a small molecule inhibitor. The structure reveals both similarities and differences between IRAK1 and IRAK4 and is suggestive of approaches to develop IRAK1- or IRAK4-specific inhibitors for potential therapeutic applications. While the IRAK4 kinase domain is capable of homodimerization in the unphosphorylated state, we found that the IRAK1 kinase domain is constitutively monomeric regardless of its phosphorylation state. Additionally, the IRAK1 kinase domain forms heterodimers with the phosphorylated, but not unphosphorylated, IRAK4 kinase domain. Collectively, these data indicate a two-step kinase activation process in which the IRAK4 kinase domain first homodimerizes in the Myddosome, leading to its trans-autophosphorylation and activation. The phosphorylated IRAK4 kinase domain then forms heterodimers with the IRAK1 kinase domain within the Myddosome, leading to its subsequent phosphorylation and activation.


Journal of Clinical Investigation | 2018

Specific covalent inhibition of MALT1 paracaspase suppresses B cell lymphoma growth

Lorena Fontan; Qi Qiao; John M. Hatcher; Gabriella Casalena; Ilkay Us; Matt Teater; Matthew Durant; Guangyan Du; Min Xia; Natalia Bilchuk; Spandan Chennamadhavuni; Giuseppe Palladino; Giorgio Inghirami; Ulrike Philippar; Hao Wu; David Scott; Nathanael S. Gray; Ari Melnick

The paracaspase MALT1 plays an essential role in activated B cell–like diffuse large B cell lymphoma (ABC DLBCL) downstream of B cell and TLR pathway genes mutated in these tumors. Although MALT1 is considered a compelling therapeutic target, the development of tractable and specific MALT1 protease inhibitors has thus far been elusive. Here, we developed a target engagement assay that provides a quantitative readout for specific MALT1-inhibitory effects in living cells. This enabled a structure-guided medicinal chemistry effort culminating in the discovery of pharmacologically tractable, irreversible substrate-mimetic compounds that bind the MALT1 active site. We confirmed that MALT1 targeting with compound 3 is effective at suppressing ABC DLBCL cells in vitro and in vivo. We show that a reduction in serum IL-10 levels exquisitely correlates with the drug pharmacokinetics and degree of MALT1 inhibition in vitro and in vivo and could constitute a useful pharmacodynamic biomarker to evaluate these compounds in clinical trials. Compound 3 revealed insights into the biology of MALT1 in ABC DLBCL, such as the role of MALT1 in driving JAK/STAT signaling and suppressing the type I IFN response and MHC class II expression, suggesting that MALT1 inhibition could prime lymphomas for immune recognition by cytotoxic immune cells.

Collaboration


Dive into the Qi Qiao's collaboration.

Top Co-Authors

Avatar

Hao Wu

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Li Wang

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Chenghua Yang

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge