Network


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

Hotspot


Dive into the research topics where Cong Xu is active.

Publication


Featured researches published by Cong Xu.


Nature Neuroscience | 2010

The serine hydrolase ABHD6 controls the accumulation and efficacy of 2-AG at cannabinoid receptors

William R. Marrs; Jacqueline L. Blankman; Eric A. Horne; Aurore Thomazeau; Yi Hsing Lin; Jonathan Coy; Ágnes L. Bodor; Giulio G. Muccioli; Sherry Shu Jung Hu; Grace Woodruff; Susan Fung; Mathieu Lafourcade; Jessica P. Alexander; Jonathan Z. Long; Weiwei Li; Cong Xu; Thomas Möller; Ken Mackie; Olivier J. Manzoni; Benjamin F. Cravatt; Nephi Stella

The endocannabinoid 2-arachidonoylglycerol (2-AG) regulates neurotransmission and neuroinflammation by activating CB1 cannabinoid receptors on neurons and CB2 cannabinoid receptors on microglia. Enzymes that hydrolyze 2-AG, such as monoacylglycerol lipase, regulate the accumulation and efficacy of 2-AG at cannabinoid receptors. We found that the recently described serine hydrolase α-β-hydrolase domain 6 (ABHD6) also controls the accumulation and efficacy of 2-AG at cannabinoid receptors. In cells from the BV-2 microglia cell line, ABHD6 knockdown reduced hydrolysis of 2-AG and increased the efficacy with which 2-AG can stimulate CB2-mediated cell migration. ABHD6 was expressed by neurons in primary culture and its inhibition led to activity-dependent accumulation of 2-AG. In adult mouse cortex, ABHD6 was located postsynaptically and its selective inhibition allowed the induction of CB1-dependent long-term depression by otherwise subthreshold stimulation. Our results indicate that ABHD6 is a rate-limiting step of 2-AG signaling and is therefore a bona fide member of the endocannabinoid signaling system.


The Journal of Neuroscience | 2007

Identification of a novel endocannabinoid-hydrolyzing enzyme expressed by microglial cells

Giulio G. Muccioli; Cong Xu; Emma Odah; Eiron Cudaback; José Cisneros; Didier M. Lambert; Maria Luz Lopez Rodriguez; Sandra M. Bajjalieh; Nephi Stella

The endocannabinoids (eCBs) anandamide and 2-arachidonoyl glycerol (2-AG) are inactivated by a two-step mechanism. First, they are carried into cells, and then anandamide is hydrolyzed by fatty acid amide hydrolase (FAAH) and 2-AG by monoacylglycerol lipase (MGL). Here we provide evidence for a previously undescribed MGL activity expressed by microglial cells. We found that the mouse microglial cell line BV-2 does not express MGL mRNA and yet efficiently hydrolyzes 2-AG. URB597 (3′-carbamoyl-biphenyl-3-yl-cyclohexylcarbamate) reduces this hydrolysis by 50%, suggesting the involvement of FAAH. The remaining activity is blocked by classic MGL inhibitors [[1,1-biphenyl]-3-yl-carbamic acid, cyclohexyl ester (URB602) and MAFP (methylarachidonyl fluorophosphate)] and is unaffected by inhibitors of COXs (cyclooxygenases), LOXs (lipooxygenases), and DGLs (diacylglycerol lipases), indicating the involvement of a novel MGL activity. Accordingly, URB602 leads to selective accumulation of 2-AG in intact BV-2 cells. Although MGL expressed in neurons is equally distributed between the cytosolic, mitochondrial, and nuclear fractions, the novel MGL activity expressed by BV-2 cells is enriched in mitochondrial and nuclear fractions. A screen for novel inhibitors of eCB hydrolysis identified several compounds that differentially block MGL, FAAH, and the novel MGL activity. Finally, we provide evidence for expression of the novel MGL by mouse primary microglia in culture. Our results suggest the presence of a novel, pharmacologically distinct, MGL activity that controls 2-AG levels in microglia.


Bioorganic & Medicinal Chemistry Letters | 2009

Synthesis and characterization of a peripherally restricted CB1 cannabinoid antagonist, URB447, that reduces feeding and body-weight gain in mice

Jesse LoVerme; Andrea Duranti; Andrea Tontini; Gilberto Spadoni; Marco Mor; Silvia Rivara; Nephi Stella; Cong Xu; Giorgio Tarzia; Daniele Piomelli

Cannabinoid CB(1) receptor antagonists reduce body weight in rodents and humans, but their clinical utility as anti-obesity agents is limited by centrally mediated side effects. Here, we describe the first mixed CB(1) antagonist/CB(2) agonist, URB447 ([4-amino-1-(4-chlorobenzyl)-2-methyl-5-phenyl-1H-pyrrol-3-yl](phenyl)methanone), which lowers food intake and body-weight gain in mice without entering the brain or antagonizing central CB(1)-dependent responses. URB447 may provide a useful pharmacological tool for investigating the cannabinoid system, and might serve as a starting point for developing clinically viable CB(1) antagonists devoid of central side effects.


Journal of Biological Chemistry | 2011

Dual inhibition of alpha/beta-hydrolase domain 6 and fatty acid amide hydrolase increases endocannabinoid levels in neurons

William R. Marrs; Eric A. Horne; Silvia Ortega-Gutiérrez; José Cisneros; Cong Xu; Yi Hsing Lin; Giulio G. Muccioli; María L. López-Rodríguez; Nephi Stella

Agonists at cannabinoid receptors, such as the phytocannabinoid Δ9-tetrahydrocannabinol, exert a remarkable array of therapeutic effects but are also associated with undesirable psychoactive side effects. Conversely, targeting enzymes that hydrolyze endocannabinoids (eCBs) allows for more precise fine-tuning of cannabinoid receptor signaling, thus providing therapeutic relief with reduced side effects. Here, we report the development and characterization of an inhibitor of eCB hydrolysis, UCM710, which augments both N-arachidonoylethanolamine and 2-arachidonoylglycerol levels in neurons. This compound displays a unique pharmacological profile in that it inhibits fatty acid amide hydrolase and α/β-hydrolase domain 6 but not monoacylglycerol lipase. Thus, UCM710 represents a novel tool to delineate the therapeutic potential of compounds that manipulate a subset of enzymes that control eCB signaling.


Biomaterials | 2015

A novel near-infrared fluorescence imaging probe that preferentially binds to cannabinoid receptors CB2R over CB1R

Xiaoxi Ling; Shaojuan Zhang; Pin Shao; Weixia Li; Ling Yang; Ying Ding; Cong Xu; Nephi Stella; Mingfeng Bai

The type 2 cannabinoid receptors (CB2R) have gained much attention recently due to their important regulatory role in a host of pathophysiological processes. However, the exact biological function of CB2R and how this function might change depending on disease progression remains unclear and could be better studied with highly sensitive and selective imaging tools for identifying the receptors. Here we report the first near infrared fluorescence imaging probe (NIR760-XLP6) that binds preferentially to CB2R over the type 1 cannabinoid receptors (CB1R). The selectivity of the probe was demonstrated by fluorescence microscopy using DBT-CB2 and DBT-CB1 cells. Furthermore, in mouse tumor models, NIR760-XLP6 showed significantly higher uptake in DBT-CB2 than that in DBT-CB1 tumors. These findings indicate that NIR760-XLP6 is a promising imaging tool for the study of CB2R regulation.


PLOS ONE | 2009

Binding of NIR-conPK and NIR-6T to Astrocytomas and Microglial Cells: Evidence for a Protein Related to TSPO

Michelle Sexton; Grace Woodruff; Eiron Cudaback; Faith R. Kreitzer; Cong Xu; Yi Hsing Lin; Thomas Möller; Mingfeng Bai; H. Charles Manning; Darryl J. Bornhop; Nephi Stella

PK 11195 and DAA1106 bind with high-affinity to the translocator protein (TSPO, formerly known as the peripheral benzodiazepine receptor). TSPO expression in glial cells increases in response to cytokines and pathological stimuli. Accordingly, [11C]-PK 11195 and [11C]-DAA1106 are recognized molecular imaging (MI) agents capable of monitoring changes in TSPO expression occurring in vivo and in response to various neuropathologies. Here we tested the pharmacological characteristics and TSPO-monitoring potential of two novel MI agents: NIR-conPK and NIR-6T. NIR-conPK is an analogue of PK 11195 conjugated to the near-infrared (NIR) emitting fluorophore: IRDye 800CW. NIR-6T is a DAA1106 analogue also conjugated to IRDye 800CW. We found that NIR-6T competed for [3H]-PK 11195 binding in astrocytoma cell homogenates with nanomolar affinity, but did not exhibit specific binding in intact astrocytoma cells in culture, indicating that NIR-6T is unlikely to constitute a useful MI agent for monitoring TSPO expression in intact cells. Conversely, we found that NIR-conPK did not compete for [3H]-PK 11195 binding in astrocytoma cell homogenate, but exhibited specific binding in intact astrocytoma cells in culture with nanomolar affinity, suggesting that NIR-conPK binds to a protein distinct, but related to, TSPO. Accordingly, treating intact astrocytoma cells and microglia in culture with cytokines led to significant changes in the amount of NIR-conPK specific binding without corresponding change in TSPO expression. Remarkably, the cytokine-induced changes in the protein targeted by NIR-conPK in intact microglia were selective, since IFN-γ (but not TNFα and TGFβ) increased the amount of NIR-conPK specific binding in these cells. Together these results suggest that NIR-conPK binds to a protein that is related to TSPO, and expressed by astrocytomas and microglia. Our results also suggest that the expression of this protein is increased by specific cytokines, and thus allows for the monitoring of a particular subtype of microglia activation.


Glia | 2015

Alkylindole-sensitive receptors modulate microglial cell migration and proliferation

Susan Fung; Allison E. Cherry; Cong Xu; Nephi Stella

Ligands targeting G protein‐coupled receptors (GPCR) expressed by microglia have been shown to regulate distinct components of their activation process, including cell proliferation, migration and differentiation into M1 or M2 phenotypes. Cannabinoids, including the active component of the Cannabis plant, tetrahydrocannabinol (THC), and the synthetic alkylindole (AI) compound, WIN55212‐2 (WIN‐2), activate two molecularly identified GPCRs: CB1 and CB2. Previous studies reported that WIN‐2 activates an additional unknown GPCR that is not activated by plant‐derived cannabinoids, and evidence indicates that microglia express these receptors. Detailed studies on the role of AI‐sensitive receptors in microglial cell activation were difficult as no selective pharmacological tools were available. Here, three newly‐developed AI analogues allowed us to determine if microglia express AI‐sensitive receptors and if so, study how they regulate the microglial cell activation process. We found that mouse microglia in primary culture express functional AI‐sensitive receptors as measured by radioligand binding and changes in intracellular cAMP levels, and that these receptors control both basal and ATP‐stimulated migration. AI analogues inhibit cell proliferation stimulated by macrophage‐colony stimulating factor (M‐CSF) without affecting basal cell proliferation. Remarkably, AI analogues do not control the expression of effector proteins characteristic of M1 or M2 phenotypes; yet activating microglia with M1 and M2 cytokines reduces the microglial response to AI analogues. Our results suggest that microglia express functional AI‐sensitive receptors that control select components of their activation process. Agonists of these novel targets might represent a novel class of therapeutics to influence the microglial cell activation process. GLIA 2015;63:1797–1808


Molecular Cancer Therapeutics | 2016

ST-11: A New Brain-Penetrant Microtubule-Destabilizing Agent with Therapeutic Potential for Glioblastoma Multiforme

Allison E. Cherry; Brian Haas; Alipi V. Naydenov; Susan Fung; Cong Xu; Katie Swinney; Michael Wagenbach; Jennifer P. Freeling; David A. Canton; Jonathan Coy; Eric A. Horne; Barry H. Rickman; Juan Jesus Vicente; John D. Scott; Rodney J. Y. Ho; Denny Liggitt; Linda Wordeman; Nephi Stella

Glioblastoma multiforme is a devastating and intractable type of cancer. Current antineoplastic drugs do not improve the median survival of patients diagnosed with glioblastoma multiforme beyond 14 to 15 months, in part because the blood–brain barrier is generally impermeable to many therapeutic agents. Drugs that target microtubules (MT) have shown remarkable efficacy in a variety of cancers, yet their use as glioblastoma multiforme treatments has also been hindered by the scarcity of brain-penetrant MT-targeting compounds. We have discovered a new alkylindole compound, ST-11, that acts directly on MTs and rapidly attenuates their rate of assembly. Accordingly, ST-11 arrests glioblastoma multiforme cells in prometaphase and triggers apoptosis. In vivo analyses reveal that unlike current antitubulin agents, ST-11 readily crosses the blood–brain barrier. Further investigation in a syngeneic orthotopic mouse model of glioblastoma multiforme shows that ST-11 activates caspase-3 in tumors to reduce tumor volume without overt toxicity. Thus, ST-11 represents the first member of a new class of brain-penetrant antitubulin therapeutic agents. Mol Cancer Ther; 15(9); 2018–29. ©2016 AACR.


Pharmacological Research | 2017

Novel indole-based compounds that differentiate alkylindole-sensitive receptors from cannabinoid receptors and microtubules: Characterization of their activity on glioma cell migration

Susan Fung; Cong Xu; Ernest Hamel; James Wager-Miller; Grace Woodruff; Aaron W. Miller; Christina A. Sanford; Ken Mackie; Nephi Stella

Graphical abstract Figure. No caption available. ABSTRACT Indole‐based compounds, such as the alkyl‐indole (AI) compound WIN55212‐2, activate the cannabinoid receptors, CB1 and CB2, two well‐characterized G protein‐coupled receptors (GPCR). Reports indicate that several indole‐based cannabinoid agonists, including WIN55212‐2, lack selectivity and interact with at least two additional targets: AI‐sensitive GPCRs and microtubules. Studying how indole‐based compounds modulate the activity of these 4 targets has been difficult as selective chemical tools were not available. Here we report the pharmacological characterization of six newly‐developed indole‐based compounds (ST‐11, ST‐23, ST‐25, ST‐29, ST‐47 and ST‐48) that exhibit distinct binding affinities at AI‐sensitive receptors, cannabinoid CB1 and CB2 receptors and the colchicine site of tubulin. Several compounds exhibit some level of selectivity for AI‐sensitive receptors, including ST‐11 that binds AI‐sensitive receptors with a Kd of 52 nM and appears to have a weaker affinity for the colchicine site of tubulin (Kd = 3.2 &mgr;M) and does not bind CB1/CB2 receptors. Leveraging these characteristics, we show that activation of AI‐sensitive receptors with ST‐11 inhibits both the basal and stimulated migration of the Delayed Brain Tumor (DBT) mouse glioma cell line. Our study describes a new series of indole‐based compounds that enable the pharmacological and functional differentiation of alkylindole‐sensitive receptors from cannabinoid receptors and microtubules.


European Journal of Medicinal Chemistry | 2018

Modified carbazoles destabilize microtubules and kill glioblastoma multiform cells

Philippe Diaz; Eric Horne; Cong Xu; Ernest Hamel; Michael Wagenbach; Ravil R. Petrov; Benjamin Uhlenbruck; Brian Haas; Parvinder Hothi; Linda Wordeman; Rick Gussio; Nephi Stella

Small molecules that target microtubules (MTs) represent promising therapeutics to treat certain types of cancer, including glioblastoma multiform (GBM). We synthesized modified carbazoles and evaluated their antitumor activity in GBM cells in culture. Modified carbazoles with an ethyl moiety linked to the nitrogen of the carbazole and a carbonyl moiety linked to distinct biaromatic rings exhibited remarkably different killing activities in human GBM cell lines and patient-derived GBM cells, with IC50 values from 67 to >10,000 nM. Measures of the activity of modified carbazoles with tubulin and microtubules coupled to molecular docking studies show that these compounds bind to the colchicine site of tubulin in a unique low interaction space that inhibits tubulin assembly. The modified carbazoles reported here represent novel chemical tools to better understand how small molecules disrupt MT functions and kill devastating cancers such as GBM.

Collaboration


Dive into the Cong Xu's collaboration.

Top Co-Authors

Avatar

Nephi Stella

University of Washington

View shared research outputs
Top Co-Authors

Avatar

Susan Fung

University of Washington

View shared research outputs
Top Co-Authors

Avatar

Eric A. Horne

University of Washington

View shared research outputs
Top Co-Authors

Avatar

Giulio G. Muccioli

Université catholique de Louvain

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Brian Haas

University of Washington

View shared research outputs
Top Co-Authors

Avatar

Grace Woodruff

University of Washington

View shared research outputs
Top Co-Authors

Avatar

Jonathan Coy

University of Washington

View shared research outputs
Top Co-Authors

Avatar

Linda Wordeman

University of Washington

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge