Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 198
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 180(4): 655-665.e18, 2020 02 20.
Article in English | MEDLINE | ID: mdl-32004463

ABSTRACT

Human endocannabinoid systems modulate multiple physiological processes mainly through the activation of cannabinoid receptors CB1 and CB2. Their high sequence similarity, low agonist selectivity, and lack of activation and G protein-coupling knowledge have hindered the development of therapeutic applications. Importantly, missing structural information has significantly held back the development of promising CB2-selective agonist drugs for treating inflammatory and neuropathic pain without the psychoactivity of CB1. Here, we report the cryoelectron microscopy structures of synthetic cannabinoid-bound CB2 and CB1 in complex with Gi, as well as agonist-bound CB2 crystal structure. Of important scientific and therapeutic benefit, our results reveal a diverse activation and signaling mechanism, the structural basis of CB2-selective agonists design, and the unexpected interaction of cholesterol with CB1, suggestive of its endogenous allosteric modulating role.


Subject(s)
Cannabinoid Receptor Agonists/pharmacology , GTP-Binding Protein alpha Subunits, Gi-Go/chemistry , Receptor, Cannabinoid, CB1/chemistry , Receptor, Cannabinoid, CB2/chemistry , Signal Transduction , Allosteric Regulation , Allosteric Site , Animals , CHO Cells , Cannabinoid Receptor Agonists/chemistry , Cannabinoids/chemistry , Cannabinoids/pharmacology , Cell Line, Tumor , Cholesterol/chemistry , Cholesterol/pharmacology , Cricetinae , Cricetulus , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Humans , Molecular Dynamics Simulation , Receptor, Cannabinoid, CB1/metabolism , Receptor, Cannabinoid, CB2/metabolism , Sf9 Cells , Spodoptera
2.
Cell ; 176(3): 459-467.e13, 2019 01 24.
Article in English | MEDLINE | ID: mdl-30639103

ABSTRACT

The cannabinoid receptor CB2 is predominately expressed in the immune system, and selective modulation of CB2 without the psychoactivity of CB1 has therapeutic potential in inflammatory, fibrotic, and neurodegenerative diseases. Here, we report the crystal structure of human CB2 in complex with a rationally designed antagonist, AM10257, at 2.8 Å resolution. The CB2-AM10257 structure reveals a distinctly different binding pose compared with CB1. However, the extracellular portion of the antagonist-bound CB2 shares a high degree of conformational similarity with the agonist-bound CB1, which led to the discovery of AM10257's unexpected opposing functional profile of CB2 antagonism versus CB1 agonism. Further structural analysis using mutagenesis studies and molecular docking revealed the molecular basis of their function and selectivity for CB2 and CB1. Additional analyses of our designed antagonist and agonist pairs provide important insight into the activation mechanism of CB2. The present findings should facilitate rational drug design toward precise modulation of the endocannabinoid system.


Subject(s)
Receptor, Cannabinoid, CB2/metabolism , Receptor, Cannabinoid, CB2/ultrastructure , Animals , Cannabinoid Receptor Antagonists/pharmacology , Cannabinoids/pharmacology , Drug Design , Endocannabinoids , Humans , Ligands , Molecular Docking Simulation , Protein Binding , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptor, Cannabinoid, CB2/chemistry , Receptors, Cannabinoid/chemistry , Receptors, Cannabinoid/metabolism , Receptors, Cannabinoid/ultrastructure , Receptors, G-Protein-Coupled/metabolism , Sf9 Cells , Structure-Activity Relationship
3.
Cell ; 172(4): 719-730.e14, 2018 02 08.
Article in English | MEDLINE | ID: mdl-29398112

ABSTRACT

Drugs frequently require interactions with multiple targets-via a process known as polypharmacology-to achieve their therapeutic actions. Currently, drugs targeting several serotonin receptors, including the 5-HT2C receptor, are useful for treating obesity, drug abuse, and schizophrenia. The competing challenges of developing selective 5-HT2C receptor ligands or creating drugs with a defined polypharmacological profile, especially aimed at G protein-coupled receptors (GPCRs), remain extremely difficult. Here, we solved two structures of the 5-HT2C receptor in complex with the highly promiscuous agonist ergotamine and the 5-HT2A-C receptor-selective inverse agonist ritanserin at resolutions of 3.0 Å and 2.7 Å, respectively. We analyzed their respective binding poses to provide mechanistic insights into their receptor recognition and opposing pharmacological actions. This study investigates the structural basis of polypharmacology at canonical GPCRs and illustrates how understanding characteristic patterns of ligand-receptor interaction and activation may ultimately facilitate drug design at multiple GPCRs.


Subject(s)
Ergotamine/chemistry , Receptor, Serotonin, 5-HT2C/chemistry , Ritanserin/chemistry , Serotonin 5-HT2 Receptor Agonists/chemistry , Serotonin 5-HT2 Receptor Antagonists/chemistry , HEK293 Cells , Humans , Obesity/drug therapy , Obesity/metabolism , Protein Domains , Receptor, Serotonin, 5-HT2C/metabolism , Schizophrenia/drug therapy , Schizophrenia/metabolism , Structure-Activity Relationship , Substance-Related Disorders/drug therapy , Substance-Related Disorders/metabolism
4.
Cell ; 167(3): 750-762.e14, 2016 Oct 20.
Article in English | MEDLINE | ID: mdl-27768894

ABSTRACT

Cannabinoid receptor 1 (CB1) is the principal target of Δ9-tetrahydrocannabinol (THC), a psychoactive chemical from Cannabis sativa with a wide range of therapeutic applications and a long history of recreational use. CB1 is activated by endocannabinoids and is a promising therapeutic target for pain management, inflammation, obesity, and substance abuse disorders. Here, we present the 2.8 Å crystal structure of human CB1 in complex with AM6538, a stabilizing antagonist, synthesized and characterized for this structural study. The structure of the CB1-AM6538 complex reveals key features of the receptor and critical interactions for antagonist binding. In combination with functional studies and molecular modeling, the structure provides insight into the binding mode of naturally occurring CB1 ligands, such as THC, and synthetic cannabinoids. This enhances our understanding of the molecular basis for the physiological functions of CB1 and provides new opportunities for the design of next-generation CB1-targeting pharmaceuticals.


Subject(s)
Cannabinoid Receptor Antagonists/chemistry , Morpholines/chemistry , Pyrazoles/chemistry , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptor, Cannabinoid, CB1/chemistry , Binding Sites , Cannabinoids/pharmacology , Cannabis/chemistry , Crystallography, X-Ray , Dronabinol/pharmacology , Endocannabinoids/pharmacology , Humans , Ligands , Morpholines/chemical synthesis , Protein Binding , Protein Conformation, alpha-Helical , Pyrazoles/chemical synthesis
5.
Nature ; 631(8020): 459-466, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38776963

ABSTRACT

Bitter taste receptors, particularly TAS2R14, play central roles in discerning a wide array of bitter substances, ranging from dietary components to pharmaceutical agents1,2. TAS2R14 is also widely expressed in extragustatory tissues, suggesting its extra roles in diverse physiological processes and potential therapeutic applications3. Here we present cryogenic electron microscopy structures of TAS2R14 in complex with aristolochic acid, flufenamic acid and compound 28.1, coupling with different G-protein subtypes. Uniquely, a cholesterol molecule is observed occupying what is typically an orthosteric site in class A G-protein-coupled receptors. The three potent agonists bind, individually, to the intracellular pockets, suggesting a distinct activation mechanism for this receptor. Comprehensive structural analysis, combined with mutagenesis and molecular dynamic simulation studies, elucidate the broad-spectrum ligand recognition and activation of the receptor by means of intricate multiple ligand-binding sites. Our study also uncovers the specific coupling modes of TAS2R14 with gustducin and Gi1 proteins. These findings should be instrumental in advancing knowledge of bitter taste perception and its broader implications in sensory biology and drug discovery.


Subject(s)
Aristolochic Acids , Cholesterol , Cryoelectron Microscopy , Receptors, G-Protein-Coupled , Taste , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/agonists , Humans , Aristolochic Acids/metabolism , Aristolochic Acids/chemistry , Aristolochic Acids/pharmacology , Cholesterol/metabolism , Ligands , Binding Sites , Molecular Dynamics Simulation , Models, Molecular , Transducin/metabolism
6.
Nature ; 585(7823): 135-140, 2020 09.
Article in English | MEDLINE | ID: mdl-32610344

ABSTRACT

Chemokines and their receptors mediate cell migration, which influences multiple fundamental biological processes and disease conditions such as inflammation and cancer1. Although ample effort has been invested into the structural investigation of the chemokine receptors and receptor-chemokine recognition2-4, less is known about endogenous chemokine-induced receptor activation and G-protein coupling. Here we present the cryo-electron microscopy structures of interleukin-8 (IL-8, also known as CXCL8)-activated human CXC chemokine receptor 2 (CXCR2) in complex with Gi protein, along with a crystal structure of CXCR2 bound to a designed allosteric antagonist. Our results reveal a unique shallow mode of binding between CXCL8 and CXCR2, and also show the interactions between CXCR2 and Gi protein. Further structural analysis of the inactive and active states of CXCR2 reveals a distinct activation process and the competitive small-molecule antagonism of chemokine receptors. In addition, our results provide insights into how a G-protein-coupled receptor is activated by an endogenous protein molecule, which will assist in the rational development of therapeutics that target the chemokine system for better pharmacological profiles.


Subject(s)
Models, Molecular , Receptors, Interleukin-8B/chemistry , Receptors, Interleukin-8B/metabolism , Signal Transduction , Allosteric Regulation , Allosteric Site , Chemokines/classification , Chemokines/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/chemistry , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Humans , Interleukin-8/metabolism , Protein Binding , Structure-Activity Relationship , Substrate Specificity
7.
Nat Immunol ; 13(12): 1155-61, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23142775

ABSTRACT

The induction of type I interferons by the bacterial secondary messengers cyclic di-GMP (c-di-GMP) or cyclic di-AMP (c-di-AMP) is dependent on a signaling axis that involves the adaptor STING, the kinase TBK1 and the transcription factor IRF3. Here we identified the heliase DDX41 as a pattern-recognition receptor (PRR) that sensed both c-di-GMP and c-di-AMP. DDX41 specifically and directly interacted with c-di-GMP. Knockdown of DDX41 via short hairpin RNA in mouse or human cells inhibited the induction of genes encoding molecules involved in the innate immune response and resulted in defective activation of STING, TBK1 and IRF3 in response to c-di-GMP or c-di-AMP. Our results suggest a mechanism whereby c-di-GMP and c-di-AMP are detected by DDX41, which forms a complex with STING to signal to TBK1-IRF3 and activate the interferon response.


Subject(s)
Cyclic GMP/analogs & derivatives , DEAD-box RNA Helicases/metabolism , Dinucleoside Phosphates/metabolism , Interferon Type I/immunology , Listeria monocytogenes/immunology , Listeria monocytogenes/metabolism , Receptors, Pattern Recognition/metabolism , Animals , Cell Line , Cyclic GMP/metabolism , DEAD-box RNA Helicases/genetics , Humans , Immunity, Innate , Interferon Regulatory Factor-3/metabolism , Macrophages/immunology , Membrane Proteins/metabolism , Mice , Protein Serine-Threonine Kinases/metabolism , RNA Interference , RNA, Small Interfering , Receptors, Pattern Recognition/genetics , Second Messenger Systems , Signal Transduction
8.
Immunity ; 40(3): 329-41, 2014 Mar 20.
Article in English | MEDLINE | ID: mdl-24560620

ABSTRACT

Stimulator of interferon genes (STING, also named MITA, MYPS, or ERIS) is an intracellular DNA sensor that induces type I interferon through its interaction with TANK-binding kinase 1 (TBK1). Here we found that the nucleotide-binding, leucine-rich-repeat-containing protein, NLRC3, reduced STING-dependent innate immune activation in response to cytosolic DNA, cyclic di-GMP (c-di-GMP), and DNA viruses. NLRC3 associated with both STING and TBK1 and impeded STING-TBK1 interaction and downstream type I interferon production. By using purified recombinant proteins, we found NLRC3 to interact directly with STING. Furthermore, NLRC3 prevented proper trafficking of STING to perinuclear and punctated region, known to be important for its activation. In animals, herpes simplex virus 1 (HSV-1)-infected Nlrc3(-/-) mice exhibited enhanced innate immunity and reduced morbidity and viral load. This demonstrates the intersection of two key pathways of innate immune regulation, NLR and STING, to fine tune host response to intracellular DNA, DNA virus, and c-di-GMP.


Subject(s)
DNA/immunology , Immunity, Innate , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Signal Transduction , Animals , Cell Line , Cyclic GMP/analogs & derivatives , Cyclic GMP/pharmacology , Cytokines/biosynthesis , Herpes Simplex/immunology , Herpes Simplex/metabolism , Herpesvirus 1, Human/physiology , Humans , Intercellular Signaling Peptides and Proteins/deficiency , Interferon Type I/biosynthesis , Mice , Mice, Knockout , Protein Binding , Protein Serine-Threonine Kinases/metabolism , Protein Transport
9.
Nature ; 546(7657): 312-315, 2017 06 08.
Article in English | MEDLINE | ID: mdl-28514449

ABSTRACT

The glucagon-like peptide-1 receptor (GLP-1R) and the glucagon receptor (GCGR) are members of the secretin-like class B family of G-protein-coupled receptors (GPCRs) and have opposing physiological roles in insulin release and glucose homeostasis. The treatment of type 2 diabetes requires positive modulation of GLP-1R to inhibit glucagon secretion and stimulate insulin secretion in a glucose-dependent manner. Here we report crystal structures of the human GLP-1R transmembrane domain in complex with two different negative allosteric modulators, PF-06372222 and NNC0640, at 2.7 and 3.0 Å resolution, respectively. The structures reveal a common binding pocket for negative allosteric modulators, present in both GLP-1R and GCGR and located outside helices V-VII near the intracellular half of the receptor. The receptor is in an inactive conformation with compounds that restrict movement of the intracellular tip of helix VI, a movement that is generally associated with activation mechanisms in class A GPCRs. Molecular modelling and mutagenesis studies indicate that agonist positive allosteric modulators target the same general region, but in a distinct sub-pocket at the interface between helices V and VI, which may facilitate the formation of an intracellular binding site that enhances G-protein coupling.


Subject(s)
Glucagon-Like Peptide-1 Receptor/chemistry , Glucagon-Like Peptide-1 Receptor/metabolism , Allosteric Regulation/drug effects , Allosteric Site/drug effects , Amino Acid Sequence , Aminopyridines/chemistry , Aminopyridines/metabolism , Aminopyridines/pharmacology , Benzamides/chemistry , Benzamides/metabolism , Benzamides/pharmacology , Crystallography, X-Ray , Glucagon-Like Peptide-1 Receptor/agonists , Humans , Models, Molecular , Phenylurea Compounds/chemistry , Phenylurea Compounds/metabolism , Phenylurea Compounds/pharmacology , Protein Domains
10.
Nature ; 547(7664): 468-471, 2017 07 27.
Article in English | MEDLINE | ID: mdl-28678776

ABSTRACT

The cannabinoid receptor 1 (CB1) is the principal target of the psychoactive constituent of marijuana, the partial agonist Δ9-tetrahydrocannabinol (Δ9-THC). Here we report two agonist-bound crystal structures of human CB1 in complex with a tetrahydrocannabinol (AM11542) and a hexahydrocannabinol (AM841) at 2.80 Å and 2.95 Å resolution, respectively. The two CB1-agonist complexes reveal important conformational changes in the overall structure, relative to the antagonist-bound state, including a 53% reduction in the volume of the ligand-binding pocket and an increase in the surface area of the G-protein-binding region. In addition, a 'twin toggle switch' of Phe2003.36 and Trp3566.48 (superscripts denote Ballesteros-Weinstein numbering) is experimentally observed and appears to be essential for receptor activation. The structures reveal important insights into the activation mechanism of CB1 and provide a molecular basis for predicting the binding modes of Δ9-THC, and endogenous and synthetic cannabinoids. The plasticity of the binding pocket of CB1 seems to be a common feature among certain class A G-protein-coupled receptors. These findings should inspire the design of chemically diverse ligands with distinct pharmacological properties.


Subject(s)
Cannabinoid Receptor Agonists/chemistry , Dronabinol/analogs & derivatives , Droperidol/analogs & derivatives , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB1/chemistry , Binding Sites , Cannabinoid Receptor Agonists/chemical synthesis , Cannabinoid Receptor Agonists/pharmacology , Crystallography, X-Ray , Dronabinol/chemical synthesis , Dronabinol/chemistry , Dronabinol/pharmacology , Droperidol/chemical synthesis , Droperidol/chemistry , Droperidol/pharmacology , Heterotrimeric GTP-Binding Proteins/metabolism , Humans , Ligands , Molecular Docking Simulation , Protein Binding , Protein Conformation , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptor, Cannabinoid, CB1/metabolism
11.
Drug Dev Res ; 84(7): 1468-1481, 2023 11.
Article in English | MEDLINE | ID: mdl-37534761

ABSTRACT

Distant metastasis is the primary reason for treatment failure in patients with nasopharyngeal carcinoma (NPC). In this study, we investigated the effect of ulinastatin (UTI) on NPC metastasis and its underlying mechanism. Highly-metastatic NPC cell lines S18 and 58F were treated with UTI and the effect on cell proliferation, migration, and invasion were determined by MTS and Transwell assays. S18 cells with luciferase-expressing (S18-1C3) were injected into the left hind footpad of nude mice to establish a model of spontaneous metastasis from the footpad to popliteal lymph node (LN). The luciferase messenger RNA (mRNA) was measured by quantitative polymerase chain reaction (qPCR), and the metastasis inhibition rate was calculated. Key molecular members of the UTI-related uPA, uPAR, and JAT/STAT3 signaling pathways were detected by qPCR and immunoblotting. UTI suppressed the migration and infiltration of S18 and 5-8F cells and suppressed the metastasis of S18 cells in vivo without affecting cell proliferation. uPAR expression decreased from 24 to 48 h after UTI treatment. The antimetastatic effect of UTI is partly due to the suppression of uPA and uPAR. UTI partially suppresses NPC metastasis by downregulating the expression of uPA and uPAR.


Subject(s)
Nasopharyngeal Neoplasms , Animals , Mice , Humans , Nasopharyngeal Carcinoma/drug therapy , Nasopharyngeal Carcinoma/metabolism , Nasopharyngeal Carcinoma/pathology , Mice, Nude , Cell Line, Tumor , Nasopharyngeal Neoplasms/drug therapy , Nasopharyngeal Neoplasms/pathology , Luciferases , Cell Movement , Neoplasm Invasiveness , Neoplasm Metastasis
12.
Genome Res ; 29(2): 193-207, 2019 02.
Article in English | MEDLINE | ID: mdl-30670628

ABSTRACT

Cell behaviors are dictated by epigenetic and transcriptional programs. Little is known about how extracellular stimuli modulate these programs to reshape gene expression and control cell behavioral responses. Here, we interrogated the epigenetic and transcriptional response of endothelial cells to VEGFA treatment and found rapid chromatin changes that mediate broad transcriptomic alterations. VEGFA-responsive genes were associated with active promoters, but changes in promoter histone marks were not tightly linked to gene expression changes. VEGFA altered transcription factor occupancy and the distal epigenetic landscape, which profoundly contributed to VEGFA-dependent changes in gene expression. Integration of gene expression, dynamic enhancer, and transcription factor occupancy changes induced by VEGFA yielded a VEGFA-regulated transcriptional regulatory network, which revealed that the small MAF transcription factors are master regulators of the VEGFA transcriptional program and angiogenesis. Collectively these results revealed that extracellular stimuli rapidly reconfigure the chromatin landscape to coordinately regulate biological responses.


Subject(s)
Epigenesis, Genetic , Neovascularization, Physiologic/genetics , Transcription, Genetic , Vascular Endothelial Growth Factor A/physiology , Animals , Cells, Cultured , Chromatin/metabolism , Enhancer Elements, Genetic , Humans , Maf Transcription Factors/metabolism , Male , Mice , Mice, Nude , Promoter Regions, Genetic , Transcription Factors/metabolism
13.
Chemistry ; 28(44): e202201388, 2022 Aug 04.
Article in English | MEDLINE | ID: mdl-35608006

ABSTRACT

Detergents are the most frequently applied reagents in membrane protein (MP) studies. The limited diversity of one-head-one-tailed traditional detergents, however, is far from sufficient for structurally distinct MPs. Expansion of detergent repertoire has a continuous momentum. In line with the speculation that detergent pre-assembly exerts superiority, herein we report for the first time cross-conjugation of two series of monomeric detergents for constructing a two-dimensional library of dimeric detergents. Optimum detergents stood out with unique preferences in the systematic evaluation of individual MPs. Furthermore, unprecedented hybrid detergents 14M8G and 14M9G enabled high-quality EM study of transporter MsbA and NMR study of G protein-coupled receptor A2A AR, respectively. Given the abundance of cross-coupling chemistries, comprehensive diversity could be readily covered that would facilitate the finding of new detergents for the manipulation of thorny MPs and innovation of the functional and structural study in future.


Subject(s)
Detergents , Membrane Proteins , Detergents/chemistry , Magnetic Resonance Spectroscopy , Membrane Proteins/chemistry , Micelles
14.
J Am Chem Soc ; 143(40): 16320-16325, 2021 10 13.
Article in English | MEDLINE | ID: mdl-34596399

ABSTRACT

Due to the lack of genetically encoded probes for fluorine-19 nuclear magnetic resonance spectroscopy (19F NMR), its utility for probing eukaryotic membrane protein dynamics is limited. Here we report an efficient method for the genetic incorporation of an unnatural amino acid (UAA), 3'-trifluoromenthyl-phenylalanine (mtfF), into cannabinoid receptor 1 (CB1) in the Baculovirus Expression System. The probe can be inserted at any environmentally sensitive site, while causing minimal structural perturbation to the target protein. Using 19F NMR and X-ray crystallography methods, we discovered that the allosteric modulator Org27569 and agonists synergistically stabilize a previously unrecognized pre-active state. An allosteric modulation model is proposed to explain Org27569's distinct behavior. We demonstrate that our site-specific 19F NMR labeling method is a powerful tool in decoding the mechanism of GPCR allosteric modulation. This new method should be broadly applicable for uncovering conformational states for many important eukaryotic membrane proteins.


Subject(s)
Indoles , Piperidines
15.
Immunity ; 36(6): 1073-86, 2012 Jun 29.
Article in English | MEDLINE | ID: mdl-22579474

ABSTRACT

STING is an essential signaling molecule for DNA and cyclic di-GMP (c-di-GMP)-mediated type I interferon (IFN) production via TANK-binding kinase 1 (TBK1) and interferon regulatory factor 3 (IRF3) pathway. It contains an N-terminal transmembrane region and a cytosolic C-terminal domain (CTD). Here, we describe crystal structures of STING CTD alone and complexed with c-di-GMP in a unique binding mode. The strictly conserved aa 153-173 region was shown to be cytosolic and participated in dimerization via hydrophobic interactions. The STING CTD functions as a dimer and the dimerization was independent of posttranslational modifications. Binding of c-di-GMP enhanced interaction of a shorter construct of STING CTD (residues 139-344) with TBK1. This suggests an extra TBK1 binding site, other than serine 358. This study provides a glimpse into the unique architecture of STING and sheds light on the mechanism of c-di-GMP-mediated TBK1 signaling.


Subject(s)
Cyclic GMP/analogs & derivatives , Membrane Proteins/chemistry , Amino Acid Sequence , Conserved Sequence , Crystallography, X-Ray , Cyclic GMP/metabolism , Dimerization , HEK293 Cells , Humans , Hydrophobic and Hydrophilic Interactions , Membrane Proteins/metabolism , Models, Molecular , Molecular Sequence Data , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Protein Binding , Protein Conformation , Protein Interaction Mapping , Protein Serine-Threonine Kinases/metabolism , Recombinant Fusion Proteins/chemistry , Sequence Alignment , Sequence Homology, Amino Acid , Structure-Activity Relationship
16.
Biochem Biophys Res Commun ; 527(3): 770-777, 2020 06 30.
Article in English | MEDLINE | ID: mdl-32446561

ABSTRACT

Nasopharyngeal carcinoma (NPC) is relatively sensitive to ionizing radiation, and radiotherapy is the main treatment modality for non-metastatic NPC. Radiation therapy generates overproduction of reactive oxygen species (ROS), which can cause DNA damage and induce apoptosis in tumors, thereby killing the malignant cells. Although dietary antioxidant supplementation reduces oxidative stress and promotes tumor progression, the effects of antioxidants on the NPC cells upon radiation have not been reported. In the present study, we showed that antioxidants (ß-Carotene, NAC, GSH) played an anti-apoptotic role in response to radiation via decreasing ROS production and inhibiting MAPK pathway in NPC cells. Based on that, we conclude that the use of supplemental antioxidants during radiotherapy should be avoided because of the possibility of tumor protection and reduced treatment efficacy.


Subject(s)
Antioxidants/pharmacology , Apoptosis/drug effects , Apoptosis/radiation effects , MAP Kinase Signaling System/drug effects , Nasopharyngeal Carcinoma/radiotherapy , Nasopharyngeal Neoplasms/radiotherapy , Cell Line, Tumor , Humans , Nasopharyngeal Carcinoma/drug therapy , Nasopharyngeal Carcinoma/metabolism , Nasopharyngeal Neoplasms/drug therapy , Nasopharyngeal Neoplasms/metabolism , Reactive Oxygen Species/metabolism
17.
FEMS Yeast Res ; 20(6)2020 09 01.
Article in English | MEDLINE | ID: mdl-32840573

ABSTRACT

Limonene, a valuable cyclic monoterpene, has been broadly studied in recent decades due to its wide application in the food, cosmetics and pharmaceutical industries. Engineering of the yeast Yarrowia lipolytica for fermentation of renewable biomass lignocellulosic hydrolysate may reduce the cost and improve the economics of bioconversion for the production of limonene. The aim of this study was to engineer Y. lipolytica to produce limonene from xylose and low-cost lignocellulosic feedstock. The heterologous genes XR and XDH and native gene XK encoding xylose assimilation enzymes, along with the heterologous genes tNDPS1 and tLS encoding orthogonal limonene biosynthetic enzymes, were introduced into the Po1f strain to facilitate xylose fermentation to limonene. The initially developed strain produced 0.44 mg/L of limonene in 72 h with 20 g/L of xylose. Overexpression of genes from the mevalonate pathway, including HMG1 and ERG12, significantly increased limonene production from xylose to ∼9.00 mg/L in 72 h. Furthermore, limonene production peaked at 20.57 mg/L with 50% hydrolysate after 72 h when detoxified lignocellulosic hydrolysate was used. This study is the first to report limonene production by yeast from lignocellulosic feedstock, and these results indicate the initial steps toward economical and sustainable production of isoprenoids from renewable biomass by engineered Y. lipolytica.


Subject(s)
Lignin/metabolism , Limonene/metabolism , Metabolic Engineering , Xylose/metabolism , Yarrowia/metabolism , Fermentation , Industrial Microbiology , Metabolic Networks and Pathways , Yarrowia/genetics
18.
Anticancer Drugs ; 31(7): 684-692, 2020 08.
Article in English | MEDLINE | ID: mdl-32282368

ABSTRACT

Quercetin, an abundant flavonoid found in various fruits and vegetables, displays multiple biological activities, including anticancer effects. Therefore, quercetin is receiving increasing attention as a potential adjuvant anticancer treatment. Gemcitabine (GEM) resistance is a major issue for clinicians and patients with advanced cancers, making it crucial to determine ways to bolster its effects. In this study, we explored the anticancer effects and mechanistic actions of quercetin in GEM-resistant cancer cells. Pancreatic cancer (BxPC-3, PANC-1) and hepatocellular carcinoma (HepG2, Huh-7) cell lines were studied. Proliferation assays showed that quercetin had cytotoxic effects on GEM-resistant cell lines (HepG2 and PANC-1), and flow cytometric analysis indicated a significant pro-apoptotic effect on these cell lines. GEM treatment, in combination with quercetin, resulted in increased anticancer effects compared with GEM alone. Quercetin led to S phase arrest in GEM-resistant cell lines, and western blot analysis revealed tumour protein p53 upregulation and cyclin D1 downregulation. This study provides mechanistic insight into the anticancer effects of quercetin and suggests that quercetin adjuvant treatment may benefit patients who are resistant to GEM therapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinoma, Hepatocellular/drug therapy , Deoxycytidine/analogs & derivatives , Liver Neoplasms/drug therapy , Pancreatic Neoplasms/drug therapy , Quercetin/pharmacology , Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/pharmacology , Apoptosis/drug effects , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cyclin D1/biosynthesis , Deoxycytidine/administration & dosage , Deoxycytidine/pharmacology , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm , Drug Synergism , Hep G2 Cells , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Quercetin/administration & dosage , Tumor Suppressor Protein p53/biosynthesis , Gemcitabine
19.
Biotechnol Lett ; 42(6): 945-956, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32090297

ABSTRACT

OBJECTIVE: Carotenoids, as potent antioxidant compounds, have gained extensive attention, especially in human health. In this study, the combination of CRISPR/Cas9 integration strategy and fermenter cultivation was utilized to obtain efficient ß-carotene-producing Yarrowia lipolytica cell factories for potential industrial application. RESULTS: The introduction of the genes of Mucor circinelloides, encoding phytoene dehydrogenase (carB) and bifunctional phytoene synthase/lycopene cyclase (carRP), contributed to the heterologous production of ß-carotene in Y. lipolytica XK2. Furthermore, ß-carotene production was efficiently enhanced by increasing the copy numbers of the carB and carRP genes and overexpressing of GGS1, ERG13, and HMG, the genes related to the mevalonate (MVA) pathway. Thus, the optimized strain overexpressed a total of eight genes, including three copies of carRP, two copies of carB, and single copies of GGS1, HMG, and ERG13. As a consequence, strain Y. lipolytica XK19 accumulated approximately 408 mg/L ß-carotene in shake flask cultures, a twenty-four-fold increase compared to the parental strain Y. lipolytica XK2. CONCLUSIONS: 4.5 g/L ß-carotene was obtained in a 5-L fermenter through a combination of genetic engineering and culture optimization, suggesting a great capacity and flexibility of Y. lipolytica in the production of carotenoids.


Subject(s)
Metabolic Engineering/methods , Yarrowia/genetics , beta Carotene/metabolism , Bioreactors , CRISPR-Cas Systems/genetics , Fermentation , Glucose/metabolism , Yarrowia/metabolism , beta Carotene/analysis
20.
Mol Pharmacol ; 96(5): 619-628, 2019 11.
Article in English | MEDLINE | ID: mdl-31515283

ABSTRACT

Cannabinoid receptor 1 (CB1) is a potential therapeutic target for the treatment of pain, obesity and obesity-related metabolic disorders, and addiction. The crystal structure of human CB1 has been determined in complex with the stabilizing antagonist AM6538. In the present study, we characterize AM6538 as a tight-binding/irreversible antagonist of CB1, as well as two derivatives of AM6538 (AM4112 and AM6542) as slowly dissociating CB1 antagonists across binding simulations and cellular signaling assays. The long-lasting nature of AM6538 was explored in vivo wherein AM6538 continues to block CP55,940-mediated behaviors in mice up to 5 days after a single injection. In contrast, the effects of SR141716A abate in mice 2 days after injection. These studies demonstrate the functional outcome of CB1 antagonist modification and open the path for development of long-lasting CB1 antagonists.


Subject(s)
Cannabinoid Receptor Antagonists/metabolism , Cannabinoid Receptor Antagonists/pharmacology , Nitrates/metabolism , Nitrates/pharmacology , Piperidines/metabolism , Piperidines/pharmacology , Pyrazoles/metabolism , Pyrazoles/pharmacology , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptor, Cannabinoid, CB1/metabolism , Animals , Binding Sites/drug effects , Binding Sites/physiology , CHO Cells , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Protein Binding/drug effects , Protein Binding/physiology , Protein Structure, Secondary , Receptor, Cannabinoid, CB1/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL