149 results on '"Karsten Melcher"'
Search Results
2. Cryo-EM structure of an activated VIP1 receptor-G protein complex revealed by a NanoBiT tethering strategy
- Author
-
Jia Duan, Dan-dan Shen, X. Edward Zhou, Peng Bi, Qiu-feng Liu, Yang-xia Tan, You-wen Zhuang, Hui-bing Zhang, Pei-yu Xu, Si-Jie Huang, Shan-shan Ma, Xin-heng He, Karsten Melcher, Yan Zhang, H. Eric Xu, and Yi Jiang
- Subjects
Science - Abstract
Vasoactive intestinal polypeptide receptor (VIP1R) is a widely expressed class B G protein-coupled receptor and a drug target for the treatment of inflammatory diseases. Here authors report a cryoelectron microscopy structure of human VIP1R bound to PACAP27 and Gs heterotrimer, which provides insights into PACAP27 binding and VIP receptor activation.
- Published
- 2020
- Full Text
- View/download PDF
3. Structure of formylpeptide receptor 2-Gi complex reveals insights into ligand recognition and signaling
- Author
-
Youwen Zhuang, Heng Liu, X. Edward Zhou, Ravi Kumar Verma, Parker W. de Waal, Wonjo Jang, Ting-Hai Xu, Lei Wang, Xing Meng, Gongpu Zhao, Yanyong Kang, Karsten Melcher, Hao Fan, Nevin A. Lambert, H. Eric Xu, and Cheng Zhang
- Subjects
Science - Abstract
Formylpeptide receptors (FPRs) are a class of chemotactic G protein-coupled receptors (GPCRs) that recognize pathogen- and host-derived formylpeptides. Here the authors report the 3.17 Å cryo-EM structure of the human FPR2-Gi signaling complex with a bound peptide agonist and in combination with computational docking and MD simulations provide mechanistic insights into formylpeptide recognition by FPRs.
- Published
- 2020
- Full Text
- View/download PDF
4. Molecular mechanisms of fentanyl mediated β-arrestin biased signaling.
- Author
-
Parker W de Waal, Jingjing Shi, Erli You, Xiaoxi Wang, Karsten Melcher, Yi Jiang, H Eric Xu, and Bradley M Dickson
- Subjects
Biology (General) ,QH301-705.5 - Abstract
The development of novel analgesics with improved safety profiles to combat the opioid epidemic represents a central question to G protein coupled receptor structural biology and pharmacology: What chemical features dictate G protein or β-arrestin signaling? Here we use adaptively biased molecular dynamics simulations to determine how fentanyl, a potent β-arrestin biased agonist, binds the μ-opioid receptor (μOR). The resulting fentanyl-bound pose provides rational insight into a wealth of historical structure-activity-relationship on its chemical scaffold. Following an in-silico derived hypothesis we found that fentanyl and the synthetic opioid peptide DAMGO require M153 to induce β-arrestin coupling, while M153 was dispensable for G protein coupling. We propose and validate an activation mechanism where the n-aniline ring of fentanyl mediates μOR β-arrestin through a novel M153 "microswitch" by synthesizing fentanyl-based derivatives that exhibit complete, clinically desirable, G protein biased coupling. Together, these results provide molecular insight into fentanyl mediated β-arrestin biased signaling and a rational framework for further optimization of fentanyl-based analgesics with improved safety profiles.
- Published
- 2020
- Full Text
- View/download PDF
5. Structural basis of Fusarium myosin I inhibition by phenamacril.
- Author
-
Yuxin Zhou, X Edward Zhou, Yuanping Gong, Yuanye Zhu, Xiaoman Cao, Joseph S Brunzelle, H Eric Xu, Mingguo Zhou, Karsten Melcher, and Feng Zhang
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Abstract
Fusarium is a genus of filamentous fungi that includes species that cause devastating diseases in major staple crops, such as wheat, maize, rice, and barley, resulting in severe yield losses and mycotoxin contamination of infected grains. Phenamacril is a novel fungicide that is considered environmentally benign due to its exceptional specificity; it inhibits the ATPase activity of the sole class I myosin of only a subset of Fusarium species including the major plant pathogens F. graminearum, F. asiaticum and F. fujikuroi. To understand the underlying mechanisms of inhibition, species specificity, and resistance mutations, we have determined the crystal structure of phenamacril-bound F. graminearum myosin I. Phenamacril binds in the actin-binding cleft in a new allosteric pocket that contains the central residue of the regulatory Switch 2 loop and that is collapsed in the structure of a myosin with closed actin-binding cleft, suggesting that pocket occupancy blocks cleft closure. We have further identified a single, transferable phenamacril-binding residue found exclusively in phenamacril-sensitive myosins to confer phenamacril selectivity.
- Published
- 2020
- Full Text
- View/download PDF
6. Bioluminescence Resonance Energy Transfer (BRET) Assay for Determination of Molecular Interactions in Living Cells
- Author
-
Kaleeckal Harikumar, Yan Yan, Ting-Hai Xu, Karsten Melcher, H. Xu, and Laurence Miller
- Subjects
Biology (General) ,QH301-705.5 - Abstract
The bioluminescence resonance energy transfer (BRET) assay can be used as an indicator of molecular approximation and/or interaction. A significant resonance energy transfer signal is generated when the acceptor, having the appropriate spectral overlap with the donor emission, is approximated with the donor. In the example provided, proteins tagged with bioluminescent Renilla luciferase (Rlu) as donor and yellow fluorescent protein (YFP) as acceptor were co-expressed in cells. This pair of donor and acceptor have an approximate Förster distance of 4.4 nm, providing the optimal working distance (Dacres et al., 2010). This technique can be used to explore the time-course of specific molecular interactions that occur in living cells.
- Published
- 2017
- Full Text
- View/download PDF
7. γ-Secretase Epsilon-cleavage Assay
- Author
-
Ting-Hai Xu, Yan Yan, Kaleeckal Harikumar, Laurence Miller, Karsten Melcher, and H. Xu
- Subjects
Biology (General) ,QH301-705.5 - Abstract
γ-Secretase epsilon-cleavage assay is derived from the cell-based Tango assay (Kang et al., 2015), and is a fast and sensitive method to determine the initial cleavage of C99 by γ-secretase. In this protocol, we use HTL cells, which are HEK293 cells with a stably integrated luciferase reporter under the control of the bacterial tetO operator element, in which C99 C terminally fused to a reversed tetracyclin-inducible activator (rTA) transcriptional activator is expressed. Endogenous or transfected γ-secretase cleaves a C terminally fused rTA transcriptional activator from C99, allowing rTA to move to the nucleus to activate a luciferase reporter gene as a measurement for γ-secretase cleavage activity.
- Published
- 2017
- Full Text
- View/download PDF
8. Detection of Membrane Protein Interactions by Cell-based Tango Assays
- Author
-
Yan Yan, Ting-Hai Xu, Kaleeckal Harikumar, Laurence Miller, Karsten Melcher, and H. Xu
- Subjects
Biology (General) ,QH301-705.5 - Abstract
The Tango assay is a protein-protein interaction assay, in which a transcription factor (rTA) is fused to a membrane-bound protein via a linker that contains a cleavage site for TEV protease, whereas a soluble interaction partner is fused to TEV protease (Barnea et al., 2008). Association between the two interaction partners leads to an efficient cleavage of the transcription factor, allowing it to translocate to the nucleus and activate a luciferase reporter gene as measurement of the interactions. In this modified assay, we fused one copy of the membrane-spanning amyloid precursor protein (APP) C99 region to TEV site-rTA (C99-TEV site-rTA) and a second copy to TEV protease (C99-TEV) to analyze intramembrane C99-C99 interaction in live cells.
- Published
- 2017
- Full Text
- View/download PDF
9. Streptavidin Bead Pulldown Assay to Determine Protein Homooligomerization
- Author
-
Ting-Hai Xu, Yan Yan, Kaleeckal Harikumar, Laurence Miller, Karsten Melcher, and H. Xu
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Pulldown assay is a conventional method to determine protein-protein interactions in vitro. Expressing a protein of interest with two different tags allows testing whether both versions can be captured via one of the two tags as homooligomeric complex. This protocol is based on streptavidin bead capture of a biotinylated protein and co-associated Flag-tagged protein using Streptavidin MagBeads.
- Published
- 2017
- Full Text
- View/download PDF
10. Structure and Physiological Regulation of AMPK
- Author
-
Yan Yan, X. Edward Zhou, H. Eric Xu, and Karsten Melcher
- Subjects
energy metabolism ,AMPK ,activation loop ,AID ,α-linker ,β-linker ,CBS ,LKB1 ,CaMKK2 ,αRIM ,Biology (General) ,QH301-705.5 ,Chemistry ,QD1-999 - Abstract
Adenosine monophosphate (AMP)-activated protein kinase (AMPK) is a heterotrimeric αβγ complex that functions as a central regulator of energy homeostasis. Energy stress manifests as a drop in the ratio of adenosine triphosphate (ATP) to AMP/ADP, which activates AMPK’s kinase activity, allowing it to upregulate ATP-generating catabolic pathways and to reduce energy-consuming catabolic pathways and cellular programs. AMPK senses the cellular energy state by competitive binding of the three adenine nucleotides AMP, ADP, and ATP to three sites in its γ subunit, each, which in turn modulates the activity of AMPK’s kinase domain in its α subunit. Our current understanding of adenine nucleotide binding and the mechanisms by which differential adenine nucleotide occupancies activate or inhibit AMPK activity has been largely informed by crystal structures of AMPK in different activity states. Here we provide an overview of AMPK structures, and how these structures, in combination with biochemical, biophysical, and mutational analyses provide insights into the mechanisms of adenine nucleotide binding and AMPK activity modulation.
- Published
- 2018
- Full Text
- View/download PDF
11. A Highly Sensitive Non-Radioactive Activity Assay for AMP-Activated Protein Kinase (AMPK)
- Author
-
Yan Yan, Xin Gu, H. Eric Xu, and Karsten Melcher
- Subjects
AMP-activated protein kinase (AMPK) ,Forkhead-associated (FHA) domain ,kinase assay ,AlphaScreen ,Biology (General) ,QH301-705.5 - Abstract
While many methods exist to quantitatively determine protein kinase activities, 32P-based radioactive assays remain the workhorse of many laboratories due to their high sensitivity, high signal to noise ratio, lack of interference by fluorescent and light-absorbing small molecules, and easy quantitation. Here, we demonstrate that the interaction between the yeast Rad53 Forkhead-associated (FHA) domain and a peptide optimized for phosphorylation by AMP-Activated Protein Kinase (AMPK), which has previously been exploited for the generation of intracellular phosphorylation sensors, can serve as a readout for a highly sensitive two-step AMPK AlphaScreen kinase assay with exceptional signal-to-noise ratio.
- Published
- 2017
- Full Text
- View/download PDF
12. H2O2 inhibits ABA-signaling protein phosphatase HAB1.
- Author
-
Madhuri Sridharamurthy, Amanda Kovach, Yang Zhao, Jian-Kang Zhu, H Eric Xu, Kunchithapadam Swaminathan, and Karsten Melcher
- Subjects
Medicine ,Science - Abstract
Due to its ability to be rapidly generated and propagated over long distances, H2O2 is an important second messenger for biotic and abiotic stress signaling in plants. In response to low water potential and high salt concentrations sensed in the roots of plants, the stress hormone abscisic acid (ABA) activates NADPH oxidase to generate H2O2, which is propagated in guard cells in leaves to induce stomatal closure and prevent water loss from transpiration. Using a reconstituted system, we demonstrate that H2O2 reversibly prevents the protein phosphatase HAB1, a key component of the core ABA-signaling pathway, from inhibiting its main target in guard cells, SnRK2.6/OST1 kinase. We have identified HAB1 C186 and C274 as H2O2-sensitive thiols and demonstrate that their oxidation inhibits both HAB1 catalytic activity and its ability to physically associate with SnRK2.6 by formation of intermolecular dimers.
- Published
- 2014
- Full Text
- View/download PDF
13. The potent Cdc7-Dbf4 (DDK) kinase inhibitor XL413 has limited activity in many cancer cell lines and discovery of potential new DDK inhibitor scaffolds.
- Author
-
Nanda Kumar Sasi, Kanchan Tiwari, Fen-Fen Soon, Dorine Bonte, Tong Wang, Karsten Melcher, H Eric Xu, and Michael Weinreich
- Subjects
Medicine ,Science - Abstract
Cdc7-Dbf4 kinase or DDK (Dbf4-dependent kinase) is required to initiate DNA replication by phosphorylating and activating the replicative Mcm2-7 DNA helicase. DDK is overexpressed in many tumor cells and is an emerging chemotherapeutic target since DDK inhibition causes apoptosis of diverse cancer cell types but not of normal cells. PHA-767491 and XL413 are among a number of potent DDK inhibitors with low nanomolar IC50 values against the purified kinase. Although XL413 is highly selective for DDK, its activity has not been extensively characterized on cell lines. We measured anti-proliferative and apoptotic effects of XL413 on a panel of tumor cell lines compared to PHA-767491, whose activity is well characterized. Both compounds were effective biochemical DDK inhibitors but surprisingly, their activities in cell lines were highly divergent. Unlike PHA-767491, XL413 had significant anti-proliferative activity against only one of the ten cell lines tested. Since XL413 did not effectively inhibit DDK in multiple cell lines, this compound likely has limited bioavailability. To identify potential leads for additional DDK inhibitors, we also tested the cross-reactivity of ∼400 known kinase inhibitors against DDK using a DDK thermal stability shift assay (TSA). We identified 11 compounds that significantly stabilized DDK. Several inhibited DDK with comparable potency to PHA-767491, including Chk1 and PKR kinase inhibitors, but had divergent chemical scaffolds from known DDK inhibitors. Taken together, these data show that several well-known kinase inhibitors cross-react with DDK and also highlight the opportunity to design additional specific, biologically active DDK inhibitors for use as chemotherapeutic agents.
- Published
- 2014
- Full Text
- View/download PDF
14. The crystal structure of the orphan nuclear receptor NR2E3/PNR ligand binding domain reveals a dimeric auto-repressed conformation.
- Author
-
M H Eileen Tan, X Edward Zhou, Fen-Fen Soon, Xiaodan Li, Jun Li, Eu-Leong Yong, Karsten Melcher, and H Eric Xu
- Subjects
Medicine ,Science - Abstract
Photoreceptor-specific nuclear receptor (PNR, NR2E3) is a key transcriptional regulator of human photoreceptor differentiation and maintenance. Mutations in the NR2E3-encoding gene cause various retinal degenerations, including Enhanced S-cone syndrome, retinitis pigmentosa, and Goldman-Favre disease. Although physiological ligands have not been identified, it is believed that binding of small molecule agonists, receptor desumoylation, and receptor heterodimerization may switch NR2E3 from a transcriptional repressor to an activator. While these features make NR2E3 a potential therapeutic target for the treatment of retinal diseases, there has been a clear lack of structural information for the receptor. Here, we report the crystal structure of the apo NR2E3 ligand binding domain (LBD) at 2.8 Å resolution. Apo NR2E3 functions as transcriptional repressor in cells and the structure of its LBD is in a dimeric auto-repressed conformation. In this conformation, the putative ligand binding pocket is filled with bulky hydrophobic residues and the activation-function-2 (AF2) helix occupies the canonical cofactor binding site. Mutations designed to disrupt either the AF2/cofactor-binding site interface or the dimer interface compromised the transcriptional repressor activity of this receptor. Together, these results reveal several conserved structural features shared by related orphan nuclear receptors, suggest that most disease-causing mutations affect the receptor's structural integrity, and allowed us to model a putative active conformation that can accommodate small ligands in its pocket.
- Published
- 2013
- Full Text
- View/download PDF
15. Abscisic acid signaling: thermal stability shift assays as tool to analyze hormone perception and signal transduction.
- Author
-
Fen-Fen Soon, Kelly M Suino-Powell, Jun Li, Eu-Leong Yong, H Eric Xu, and Karsten Melcher
- Subjects
Medicine ,Science - Abstract
Abscisic acid (ABA) is a plant hormone that plays important roles in growth and development. ABA is also the central regulator to protect plants against abiotic stresses, such as drought, high salinity, and adverse temperatures, and ABA signaling is therefore a promising biotechnological target for the generation of crops with increased stress resistance. Recently, a core signal transduction pathway has been established, in which ABA receptors, type 2C protein phosphatases, and AMPK-related protein kinases control the regulation of transcription factors, ion channels, and enzymes. Here we use a simple protein thermal stability shift assay to independently validate key aspects of this pathway and to demonstrate the usefulness of this technique to detect and characterize very weak (Kd ≥ 50 µM) interactions between receptors and physiological and synthetic agonists, to determine and analyze protein-protein interactions, and to screen small molecule inhibitors.
- Published
- 2012
- Full Text
- View/download PDF
16. Structure and Enzymatic Activity of an Intellectual Disability-Associated Ornithine Decarboxylase Variant, G84R
- Author
-
X. Edward Zhou, Chad R. Schultz, Kelly Suino Powell, Amy Henrickson, Jared Lamp, Joseph S. Brunzelle, Borries Demeler, Irving E. Vega, André S. Bachmann, and Karsten Melcher
- Subjects
General Chemical Engineering ,General Chemistry - Abstract
Ornithine decarboxylase (ODC) is a rate-limiting enzyme for the synthesis of polyamines (PAs). PAs are required for proliferation, and increased ODC activity is associated with cancer and neural over-proliferation. ODC levels and activity are therefore tightly regulated, including through the ODC-specific inhibitor, antizyme AZ1. Recently, ODC G84R has been reported as a partial loss-of-function variant that is associated with intellectual disability and seizures. However, G84 is distant from both the catalytic center and the ODC homodimerization interface. To understand how G84R modulates ODC activity, we have determined the crystal structure of ODC G84R in both the presence and the absence of the cofactor pyridoxal 5-phosphate. The structures show that the replacement of G84 by arginine leads to hydrogen bond formation of R84 with F420, the last residue of the ODC C-terminal helix, a structural element that is involved in the AZ1-mediated proteasomal degradation of ODC. In contrast, the catalytic center is essentially indistinguishable from that of wildtype ODC. We therefore reanalyzed the catalytic activity of ODC G84R and found that it is rescued when the protein is purified in the presence of a reducing agent to mimic the reducing environment of the cytoplasm. This suggests that R84 may exert its neurological effects not through reducing ODC catalytic activity but through misregulation of its AZ1-mediated proteasomal degradation.
- Published
- 2022
17. Structural basis of binding and inhibition of ornithine decarboxylase by 1-amino-oxy-3-aminopropane
- Author
-
Irving E. Vega, Bilal Aleiwi, Edmund Ellsworth, Kelly Suino-Powell, X. Edward Zhou, André S. Bachmann, Joseph S. Brunzelle, Chad R. Schultz, Jared Lamp, and Karsten Melcher
- Subjects
genetic structures ,education ,Ornithine Decarboxylase ,Biochemistry ,Article ,Cofactor ,Ornithine decarboxylase ,chemistry.chemical_compound ,Protein Domains ,mental disorders ,Humans ,Molecular Biology ,Pyridoxal ,chemistry.chemical_classification ,Propylamines ,biology ,fungi ,Substrate (chemistry) ,Cell Biology ,Ornithine Decarboxylase Inhibitors ,Ornithine ,Oxime ,Enzyme ,chemistry ,Covalent bond ,biology.protein ,psychological phenomena and processes ,Protein Binding - Abstract
Ornithine decarboxylase (ODC) is the rate-limiting enzyme for the synthesis of polyamines (PAs). PAs are oncometabolites that are required for proliferation, and pharmaceutical ODC inhibition is pursued for the treatment of hyperproliferative diseases, including cancer and infectious diseases. The most potent ODC inhibitor is 1-amino-oxy-3-aminopropane (APA). A previous crystal structure of an ODC–APA complex indicated that APA non-covalently binds ODC and its cofactor pyridoxal 5-phosphate (PLP) and functions by competing with the ODC substrate ornithine for binding to the catalytic site. We have revisited the mechanism of APA binding and ODC inhibition through a new crystal structure of APA-bound ODC, which we solved at 2.49 Å resolution. The structure unambiguously shows the presence of a covalent oxime between APA and PLP in the catalytic site, which we confirmed in solution by mass spectrometry. The stable oxime makes extensive interactions with ODC but cannot be catabolized, explaining APA's high potency in ODC inhibition. In addition, we solved an ODC/PLP complex structure with citrate bound at the substrate-binding pocket. These two structures provide new structural scaffolds for developing more efficient pharmaceutical ODC inhibitors.
- Published
- 2021
18. Structural insights into the lipid and ligand regulation of serotonin receptors
- Author
-
H. Eric Xu, Hsin-Yung Yen, Xi Cheng, Huibing Zhang, David E. Gloriam, Carol V. Robinson, Jia Guo, Yi Jiang, Kasper Harpsøe, Karsten Melcher, Sijie Huang, X. Edward Zhou, Hualiang Jiang, Yan Zhang, Ícaro Ariel Simon, Dan-Dan Shen, Peiyu Xu, Chunyou Mao, and Bo Svensson
- Subjects
Models, Molecular ,Agonist ,medicine.drug_class ,Aripiprazole ,Phospholipid ,Ligands ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Phosphatidylinositol Phosphates ,medicine ,Humans ,Phosphatidylinositol ,Receptor ,5-HT receptor ,030304 developmental biology ,0303 health sciences ,Binding Sites ,Multidisciplinary ,Chemistry ,Cryoelectron Microscopy ,Water ,Serotonin 5-HT1 Receptor Agonists ,Ligand (biochemistry) ,Heterotrimeric GTP-Binding Proteins ,Lipids ,3. Good health ,Cell biology ,Transmembrane domain ,Cholesterol ,Receptor, Serotonin, 5-HT1A ,Serotonin ,Apoproteins ,Receptors, Serotonin, 5-HT1 ,030217 neurology & neurosurgery - Abstract
Serotonin, or 5-hydroxytryptamine (5-HT), is an important neurotransmitter1,2 that activates the largest subtype family of G-protein-coupled receptors3. Drugs that target 5-HT1A, 5-HT1D, 5-HT1E and other 5-HT receptors are used to treat numerous disorders4. 5-HT receptors have high levels of basal activity and are subject to regulation by lipids, but the structural basis for the lipid regulation and basal activation of these receptors and the pan-agonism of 5-HT remains unclear. Here we report five structures of 5-HT receptor–G-protein complexes: 5-HT1A in the apo state, bound to 5-HT or bound to the antipsychotic drug aripiprazole; 5-HT1D bound to 5-HT; and 5-HT1E in complex with a 5-HT1E- and 5-HT1F-selective agonist, BRL-54443. Notably, the phospholipid phosphatidylinositol 4-phosphate is present at the G-protein–5-HT1A interface, and is able to increase 5-HT1A-mediated G-protein activity. The receptor transmembrane domain is surrounded by cholesterol molecules—particularly in the case of 5-HT1A, in which cholesterol molecules are directly involved in shaping the ligand-binding pocket that determines the specificity for aripiprazol. Within the ligand-binding pocket of apo-5-HT1A are structured water molecules that mimic 5-HT to activate the receptor. Together, our results address a long-standing question of how lipids and water molecules regulate G-protein-coupled receptors, reveal how 5-HT acts as a pan-agonist, and identify the determinants of drug recognition in 5-HT receptors. Cryo-electron microscopy structures of three different serotonin receptors in complex with serotonin and other agonists provide insights into the role of lipids in regulating these receptors and the structural basis of ligand recognition.
- Published
- 2021
19. Structure of nucleosome-bound DNA methyltransferases DNMT3A and DNMT3B
- Author
-
X. Edward Zhou, Karsten Melcher, Peter A. Jones, H. Eric Xu, Minmin Liu, Gongpu Zhao, Ting-Hai Xu, and Gangning Liang
- Subjects
0303 health sciences ,Multidisciplinary ,Methyltransferase ,Chemistry ,Protein domain ,Plasma protein binding ,Methylation ,Linker DNA ,Cell biology ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,DNA methylation ,Nucleosome ,030217 neurology & neurosurgery ,DNA ,030304 developmental biology - Abstract
CpG methylation by de novo DNA methyltransferases (DNMTs) 3A and 3B is essential for mammalian development and differentiation and is frequently dysregulated in cancer1. These two DNMTs preferentially bind to nucleosomes, yet cannot methylate the DNA wrapped around the nucleosome core2, and they favour the methylation of linker DNA at positioned nucleosomes3,4. Here we present the cryo-electron microscopy structure of a ternary complex of catalytically competent DNMT3A2, the catalytically inactive accessory subunit DNMT3B3 and a nucleosome core particle flanked by linker DNA. The catalytic-like domain of the accessory DNMT3B3 binds to the acidic patch of the nucleosome core, which orients the binding of DNMT3A2 to the linker DNA. The steric constraints of this arrangement suggest that nucleosomal DNA must be moved relative to the nucleosome core for de novo methylation to occur. Catalytically inactive DNMT3B3 is crucial in de novo CpG methylation of DNA, interacting with the nucleosome core to orient catalytically active DNMT3A2 so that it can bind to nearby linker DNA.
- Published
- 2020
20. Cryo-EM structure of an activated VIP1 receptor-G protein complex revealed by a NanoBiT tethering strategy
- Author
-
Peiyu Xu, Jia Duan, Yan Zhang, Xinheng He, Yangxia Tan, Sijie Huang, X. Edward Zhou, H. Eric Xu, Karsten Melcher, Dan-Dan Shen, Huibing Zhang, Youwen Zhuang, Qiufeng Liu, Yi Jiang, Shanshan Ma, and Peng Bi
- Subjects
0301 basic medicine ,G protein ,Cryo-electron microscopy ,Receptors, Vasoactive Intestinal Polypeptide, Type I ,Science ,Vasoactive intestinal peptide ,General Physics and Astronomy ,02 engineering and technology ,General Biochemistry, Genetics and Molecular Biology ,Article ,Receptors, G-Protein-Coupled ,03 medical and health sciences ,G protein-coupled receptors ,GTP-Binding Proteins ,Cryoelectron microscopy ,medicine ,Humans ,Receptor ,lcsh:Science ,G protein-coupled receptor ,Multidisciplinary ,Chemistry ,Drug discovery ,General Chemistry ,021001 nanoscience & nanotechnology ,Transmembrane protein ,Dynamic Light Scattering ,Cell biology ,Microscopy, Electron ,030104 developmental biology ,Mechanism of action ,Pituitary Adenylate Cyclase-Activating Polypeptide ,lcsh:Q ,medicine.symptom ,0210 nano-technology - Abstract
Vasoactive intestinal polypeptide receptor (VIP1R) is a widely expressed class B G protein-coupled receptor and a drug target for the treatment of neuronal, metabolic, and inflammatory diseases. However, our understanding of its mechanism of action and the potential of drug discovery targeting this receptor is limited by the lack of structural information of VIP1R. Here we report a cryo-electron microscopy structure of human VIP1R bound to PACAP27 and Gs heterotrimer, whose complex assembly is stabilized by a NanoBiT tethering strategy. Comparison with other class B GPCR structures reveals that PACAP27 engages VIP1R with its N-terminus inserting into the ligand binding pocket at the transmembrane bundle of the receptor, which subsequently couples to the G protein in a receptor-specific manner. This structure has provided insights into the molecular basis of PACAP27 binding and VIP receptor activation. The methodology of the NanoBiT tethering may help to provide structural information of unstable complexes., Vasoactive intestinal polypeptide receptor (VIP1R) is a widely expressed class B G protein-coupled receptor and a drug target for the treatment of inflammatory diseases. Here authors report a cryoelectron microscopy structure of human VIP1R bound to PACAP27 and Gs heterotrimer, which provides insights into PACAP27 binding and VIP receptor activation.
- Published
- 2020
21. Long-chain fatty acyl-CoA esters regulate metabolism via allosteric control of AMPK β1 isoforms
- Author
-
Kim Loh, Bruce E. Kemp, John W. Scott, Michael W. Parker, Gregory R. Steinberg, Stephen L. Pinkosky, Yan Yan, Emily A. Day, Ashfaqul Hoque, William J. Smiles, Eric M. Desjardins, Kevin R.W. Ngoei, Sandra Galic, Brennan K. Smith, Rebecca J. Ford, Naomi X.Y. Ling, Christopher G. Langendorf, Tracy L. Nero, Jonathan S. Oakhill, and Karsten Melcher
- Subjects
Male ,Models, Molecular ,Endocrinology, Diabetes and Metabolism ,Allosteric regulation ,Thiophenes ,AMP-Activated Protein Kinases ,Article ,Mice ,03 medical and health sciences ,Acyl-CoA ,chemistry.chemical_compound ,Allosteric Regulation ,AMP-activated protein kinase ,Catalytic Domain ,Physiology (medical) ,Internal Medicine ,Animals ,Phosphorylation ,Protein kinase A ,Beta oxidation ,030304 developmental biology ,0303 health sciences ,Palmitoyl Coenzyme A ,biology ,Biphenyl Compounds ,030302 biochemistry & molecular biology ,AMPK ,Esters ,Cell Biology ,Isoenzymes ,Mice, Inbred C57BL ,Malonyl-CoA ,chemistry ,Biochemistry ,Pyrones ,Mutation ,biology.protein ,lipids (amino acids, peptides, and proteins) ,Acyl Coenzyme A ,Energy source ,Oxidation-Reduction - Abstract
Long-chain fatty acids (LCFAs) play important roles in cellular energy metabolism, acting as both an important energy source and signalling molecules1. LCFA-CoA esters promote their own oxidation by acting as allosteric inhibitors of acetyl-CoA carboxylase, which reduces the production of malonyl-CoA and relieves inhibition of carnitine palmitoyl-transferase 1, thereby promoting LCFA-CoA transport into the mitochondria for β-oxidation2,3,4,5,6. Here we report a new level of regulation wherein LCFA-CoA esters per se allosterically activate AMP-activated protein kinase (AMPK) β1–containing isoforms to increase fatty acid oxidation through phosphorylation of acetyl-CoA carboxylase. Activation of AMPK by LCFA-CoA esters requires the allosteric drug and metabolite site formed between the α-subunit kinase domain and the β-subunit. β1 subunit mutations that inhibit AMPK activation by the small-molecule activator A769662, which binds to the allosteric drug and metabolite site, also inhibit activation by LCFA-CoAs. Thus, LCFA-CoA metabolites act as direct endogenous AMPK β1–selective activators and promote LCFA oxidation.
- Published
- 2020
22. Structure of formylpeptide receptor 2-Gi complex reveals insights into ligand recognition and signaling
- Author
-
Ravi Kumar Verma, Lei Wang, Heng Liu, Hao Fan, Parker W. de Waal, X. Edward Zhou, Gongpu Zhao, Karsten Melcher, H. Eric Xu, Xing Meng, Youwen Zhuang, Wonjo Jang, Yanyong Kang, Cheng Zhang, Nevin A. Lambert, and Ting-Hai Xu
- Subjects
0301 basic medicine ,Protein Conformation ,Science ,General Physics and Astronomy ,Computational biology ,Mechanism of action ,Ligands ,Molecular Docking Simulation ,Article ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,0302 clinical medicine ,Membrane proteins ,Animals ,Humans ,Receptors, Lipoxin ,Binding site ,Receptor ,lcsh:Science ,G protein-coupled receptor ,Binding Sites ,Multidisciplinary ,Chemistry ,Cryoelectron Microscopy ,Chemotaxis ,General Chemistry ,Ligand (biochemistry) ,Receptors, Formyl Peptide ,Rats ,3. Good health ,030104 developmental biology ,Docking (molecular) ,030220 oncology & carcinogenesis ,Mutation ,lcsh:Q ,Signal transduction ,Peptides ,Signal Transduction - Abstract
Formylpeptide receptors (FPRs) as G protein-coupled receptors (GPCRs) can recognize formylpeptides derived from pathogens or host cells to function in host defense and cell clearance. In addition, FPRs, especially FPR2, can also recognize other ligands with a large chemical diversity generated at different stages of inflammation to either promote or resolve inflammation in order to maintain a balanced inflammatory response. The mechanism underlying promiscuous ligand recognition and activation of FPRs is not clear. Here we report a cryo-EM structure of FPR2-Gi signaling complex with a peptide agonist. The structure reveals a widely open extracellular region with an amphiphilic environment for ligand binding. Together with computational docking and simulation, the structure suggests a molecular basis for the recognition of formylpeptides and a potential mechanism of receptor activation, and reveals conserved and divergent features in Gi coupling. Our results provide a basis for understanding the molecular mechanism of the functional promiscuity of FPRs., Formylpeptide receptors (FPRs) are a class of chemotactic G protein-coupled receptors (GPCRs) that recognize pathogen- and host-derived formylpeptides. Here the authors report the 3.17 Å cryo-EM structure of the human FPR2-Gi signaling complex with a bound peptide agonist and in combination with computational docking and MD simulations provide mechanistic insights into formylpeptide recognition by FPRs.
- Published
- 2020
23. Identification and structural insight of an effective PPARγ modulator with improved therapeutic index for anti-diabetic drug discovery
- Author
-
Haowen Jiang, Jia Li, Hui Gao, Wei Yi, Kelly Suino-Powell, Karsten Melcher, Xinwen Zhang, Lei Ma, Jingjing Shi, X. Edward Zhou, H. Eric Xu, Guanguan Zhao, Jing-Ya Li, Xiyong Yu, Zhi Zhou, and Y. Sun
- Subjects
0303 health sciences ,Chemistry ,Drug discovery ,Allosteric regulation ,Regulator ,Lipid metabolism ,General Chemistry ,Pharmacology ,Partial agonist ,03 medical and health sciences ,0302 clinical medicine ,Therapeutic index ,In vivo ,030220 oncology & carcinogenesis ,Glucose homeostasis ,030304 developmental biology - Abstract
Peroxisome proliferator-activated receptor γ (PPARγ) is a key regulator of glucose homeostasis and lipid metabolism, and an important target for the development of modern anti-diabetic drugs. However, current PPARγ-targeting anti-diabetic drugs such as classical thiazolidinediones (TZDs) are associated with undesirable side effects. To address this concern, we here describe the structure-based design, synthesis, identification and detailed in vitro and in vivo characterization of a novel, decanoic acid (DA)-based and selective PPARγ modulator (SPPARγM), VSP-77, especially (S)-VSP-77, as the potential “hit” for the development of improved and safer anti-diabetic therapeutics. We have also determined the co-crystal structure of the PPARγ ligand-binding domain (LBD) in complex with two molecules of (S)-VSP-77, which reveal a previously undisclosed allosteric binding mode. Overall, these findings not only demonstrate the therapeutic advantage of (S)-VSP-77 over current TZD drugs and representative partial agonist INT131, but also provide a rational basis for the development of future SPPARγMs as safe and highly efficacious anti-diabetic drugs.
- Published
- 2020
24. The structural basis of arrestin–GPCR interactions
- Author
-
X. Edward Zhou and Karsten Melcher
- Published
- 2022
25. Structures of the entire human opioid receptor family
- Author
-
Yue Wang, Youwen Zhuang, Jeffrey F. DiBerto, X. Edward Zhou, Gavin P. Schmitz, Qingning Yuan, Manish K. Jain, Weiyi Liu, Karsten Melcher, Yi Jiang, Bryan L. Roth, and H. Eric Xu
- Subjects
General Biochemistry, Genetics and Molecular Biology - Published
- 2023
26. Molecular recognition of morphine and fentanyl by the human μ-opioid receptor
- Author
-
Youwen Zhuang, Yue Wang, Bingqing He, Xinheng He, X. Edward Zhou, Shimeng Guo, Qidi Rao, Jiaqi Yang, Jinyu Liu, Qingtong Zhou, Xiaoxi Wang, Mingliang Liu, Weiyi Liu, Xiangrui Jiang, Dehua Yang, Hualiang Jiang, Jingshan Shen, Karsten Melcher, Hong Chen, Yi Jiang, Xi Cheng, Ming-Wei Wang, Xin Xie, and H. Eric Xu
- Subjects
Analgesics, Opioid ,Fentanyl ,Arrestin ,Morphine ,GTP-Binding Proteins ,Receptors, Opioid, mu ,Humans ,General Biochemistry, Genetics and Molecular Biology - Abstract
Morphine and fentanyl are among the most used opioid drugs that confer analgesia and unwanted side effects through both G protein and arrestin signaling pathways of μ-opioid receptor (μOR). Here, we report structures of the human μOR-G protein complexes bound to morphine and fentanyl, which uncover key differences in how they bind the receptor. We also report structures of μOR bound to TRV130, PZM21, and SR17018, which reveal preferential interactions of these agonists with TM3 side of the ligand-binding pocket rather than TM6/7 side. In contrast, morphine and fentanyl form dual interactions with both TM3 and TM6/7 regions. Mutations at the TM6/7 interface abolish arrestin recruitment of μOR promoted by morphine and fentanyl. Ligands designed to reduce TM6/7 interactions display preferential G protein signaling. Our results provide crucial insights into fentanyl recognition and signaling of μOR, which may facilitate rational design of next-generation analgesics.
- Published
- 2021
27. Crystal structure of heliorhodopsin 48C12
- Author
-
Karsten Melcher, Lu Yang, Ruixue Xia, H. Eric Xu, Yuanzheng He, Zhenmei Xu, X. Edward Zhou, Xiang Gao, Na Wang, Guangfu Wang, Yuying Shi, and Yu Leng
- Subjects
Models, Molecular ,Nanocrystallography ,Protein Conformation ,Cell Biology ,Crystal structure ,Biology ,Hormone receptors ,Crystallography, X-Ray ,Cell biology ,Hormone receptor ,Signaling proteins ,Rhodopsins, Microbial ,Protein Multimerization ,Molecular Biology ,Letter to the Editor - Published
- 2019
28. Structure and dynamics of the active human parathyroid hormone receptor-1
- Author
-
Ieva Sutkeviciute, X.E. Zhou, Karsten Melcher, Thomas J. Gardella, Alex D. White, Lisa J. Clark, C. Li, Ming-Wei Wang, Shanshan Ma, H.E. Xu, Yan Zhang, Xiaoqing Cai, Li Zhao, D.-D. Shen, Jean-Pierre Vilardaga, Yanyong Kang, Chuankui Li, Tomoyuki Watanabe, Yi Jiang, D. Yang, P.W. de Waal, Frederic Jean-Alphonse, Antao Dai, Jianmin Chen, Chinese Academy of Science (CAS), University of Chinese Academy of Sciences [Beijing] (UCAS), University of Pittsburgh School of Medicine, and Pennsylvania Commonwealth System of Higher Education (PCSHE)
- Subjects
G protein ,Amino Acid Motifs ,Protein domain ,Parathyroid hormone ,Plasma protein binding ,Article ,[SDV.BDLR.RS]Life Sciences [q-bio]/Reproductive Biology/Sexual reproduction ,03 medical and health sciences ,0302 clinical medicine ,Protein Domains ,[SDV.BC.IC]Life Sciences [q-bio]/Cellular Biology/Cell Behavior [q-bio.CB] ,Humans ,Receptor ,Receptor, Parathyroid Hormone, Type 1 ,030304 developmental biology ,0303 health sciences ,Multidisciplinary ,Chemistry ,Cryoelectron Microscopy ,[SDV.BBM.BM]Life Sciences [q-bio]/Biochemistry, Molecular Biology/Molecular biology ,[SDV.BBM.MN]Life Sciences [q-bio]/Biochemistry, Molecular Biology/Molecular Networks [q-bio.MN] ,Transmembrane domain ,Parathyroid Hormone ,Hormone receptor ,Helix ,[SDV.SP.PHARMA]Life Sciences [q-bio]/Pharmaceutical sciences/Pharmacology ,Biophysics ,030217 neurology & neurosurgery ,Protein Binding - Abstract
Bone-cell regulation, fleshed out One of many medically relevant G protein–coupled receptors, parathyroid hormone receptor-1 (PTH1R) functions in the control of calcium homeostasis and bone physiology. Zhao et al. used cryo–election microscopy to observe the structure of PTH1R in a complex with a modified form of parathyroid hormone and stimulatory G protein. The structural model helps explain how parathyroid hormone interacts with its receptor and the molecular basis for receptor activation. Science , this issue p. 148
- Published
- 2019
29. Development of highly potent glucocorticoids for steroid-resistant severe asthma
- Author
-
Erli You, Sok Kean Khoo, Zhijian Xi, Wu Zhongshan, Yuanzheng He, Hongbo Liu, Booki Min, Karsten Melcher, Quang Tam Nguyen, Jianshuang Li, Wenli Qiu, Jingjing Shi, Sun Feng, Tao Yang, Xiaozhu Huang, Wei Yi, Xin Ren, and H. Eric Xu
- Subjects
Male ,Inflammation ,Pharmacology ,Severity of Illness Index ,Fluticasone propionate ,Mice ,Receptors, Glucocorticoid ,Glucocorticoid receptor ,Drug Development ,medicine ,Animals ,Anti-Asthmatic Agents ,Glucocorticoids ,Dexamethasone ,Asthma ,Multidisciplinary ,Lung ,Inhalation ,business.industry ,Biological Sciences ,medicine.disease ,Disease Models, Animal ,medicine.anatomical_structure ,Female ,medicine.symptom ,business ,hormones, hormone substitutes, and hormone antagonists ,Glucocorticoid ,medicine.drug - Abstract
Clinical application of inhaled glucocorticoids (GCs) has been hampered in the case of steroid-resistant severe asthma. To overcome this limitation, we have developed a series of highly potent GCs, including VSGC12, VSG158, and VSG159 based on the structural insight into the glucocorticoid receptor (GR). Particularly, VSG158 exhibits a maximal repression of lung inflammation and is 10 times more potent than the currently most potent clinical GC, Fluticasone Furoate (FF), in a murine model of asthma. More importantly, VSG158 displays a unique property to reduce neutrophilic inflammation in a steroid-resistant airway inflammation model, which is refractory to clinically available GCs, including dexamethasone and FF. VSG158 and VSG159 are able to deliver effective treatments with reduced off-target and side effects. In addition, these GCs also display pharmacokinetic properties that are suitable for the inhalation delivery method for asthma treatment. Taken together, the excellent therapeutic and side-effect profile of these highly potent GCs holds promise for treating steroid-resistant severe asthma.
- Published
- 2019
30. Structures of AMP-activated protein kinase bound to novel pharmacological activators in phosphorylated, non-phosphorylated, and nucleotide-free states
- Author
-
Yan Yan, Joseph S. Brunzelle, Y. Li, X.E. Zhou, Patrick R. Griffin, Karsten Melcher, Y. Hitoshi, H.E. Xu, Simon J. Shaw, and Scott J. Novick
- Subjects
0301 basic medicine ,Enzyme Activators ,AMP-Activated Protein Kinases ,Biochemistry ,Dephosphorylation ,03 medical and health sciences ,AMP-activated protein kinase ,Catalytic Domain ,Humans ,Transferase ,Kinase activity ,Protein kinase A ,Molecular Biology ,030102 biochemistry & molecular biology ,biology ,Chemistry ,AMPK ,Hep G2 Cells ,Cell Biology ,Recombinant Proteins ,Cell biology ,030104 developmental biology ,Protein kinase domain ,biology.protein ,Phosphorylation ,Signal Transduction - Abstract
AMP-activated protein kinase (AMPK) is an attractive therapeutic target for managing metabolic diseases. A class of pharmacological activators, including Merck 991, binds the AMPK ADaM site, which forms the interaction surface between the kinase domain (KD) of the α-subunit and the carbohydrate-binding module (CBM) of the β-subunit. Here, we report the development of two new 991-derivative compounds, R734 and R739, which potently activate AMPK in a variety of cell types, including β(2)-specific skeletal muscle cells. Surprisingly, we found that they have only minor effects on direct kinase activity of the recombinant α(1)β(2)γ(1) isoform yet robustly enhance protection against activation loop dephosphorylation. This mode of activation is reminiscent of that of ADP, which activates AMPK by binding to the nucleotide-binding sites in the γ-subunit, more than 60 Å away from the ADaM site. To understand the mechanisms of full and partial AMPK activation, we determined the crystal structures of fully active phosphorylated AMPK α(1)β(1)γ(1) bound to AMP and R734/R739 as well as partially active nonphosphorylated AMPK bound to R734 and AMP and phosphorylated AMPK bound to R734 in the absence of added nucleotides at
- Published
- 2019
31. Generation of a New Frizzled 2 Flox Mouse Model to Clarify Its Role in Development
- Author
-
Jarrett Adams, Zhendong A. Zhong, Bart O. Williams, Megan N. Michalski, Transgenics Core, Stephane Angers, Karsten Melcher, Sachdev S. Sidhu, Cassandra R. Diegel, Ian Beddows, Kelly Suino-Powell, Levi L. Blazer, and Vai Vivarium
- Subjects
FZD1 ,Knockout mouse ,CRISPR ,Cre recombinase ,Computational biology ,Biology ,Homologous recombination ,Null allele ,Phenotype ,Gene knockout - Abstract
It is currently accepted that Wnt receptors, Frizzleds (Fzd), have high functional redundancy, making individual receptors challenging to target therapeutically. Specifically, Fzd2 is believed to be functionally redundant with Fzd1 and Fzd7, findings which were based largely on previously published global knockout mouse studies. Conversely, a Fzd2 global knockout mouse model developed by the International Mouse Phenotype Consortium (IMPC) is early embryonic lethal, suggesting Fzd2 is critical for early embryonic development. If global deletion of Fzd2 leads to early lethality, floxed models are necessary to identify tissue-specific phenotypes. We found that a previously published Fzd2 flox model does not fully delete Fzd2 function. To reconcile the contradictory findings in Fzd2 mouse models and allow for tissue-specific studies of Fzd2, we have generated a new flox model using a modified two-cell homologous recombination CRISPR approach. We demonstrated successful simultaneous insertion of two loxP sites fully surrounding the Fzd2 gene and confirmed cre-mediated recombination deletes the sequence between the loxP sites leading to a Fzd2 null allele. Preliminary studies suggest global knockouts are early embryonic lethal and full characterization of the tissue-specific effects of Fzd2 deletion is currently underway. This work suggests Fzd2 uniquely regulates development and emphasizes the importance of thorough validation of newly generated mouse models.
- Published
- 2021
32. Dopaminergic Pharmacology Revealed by Structures of Human Dopamine D3 Receptor-G i Complexes
- Author
-
Yangxia Tan, Dan-Dan Shen, Xi Cheng, Karsten Melcher, Yi Jiang, Chunyou Mao, Sijie Huang, Yan Zhang, Hualiang Jiang, Chenjun Jia, Peiyu Xu, Xuekui Yu, X. Edward Zhou, and H. Eric Xu
- Subjects
Agonist ,History ,Pramipexole ,Polymers and Plastics ,Chemistry ,medicine.drug_class ,Dopaminergic ,Industrial and Manufacturing Engineering ,Cell biology ,Dopamine receptor ,Dopamine receptor D3 ,Dopamine ,medicine ,Homology modeling ,Business and International Management ,G protein-coupled receptor ,medicine.drug - Abstract
The dopamine system, including five dopamine receptors (D1R to D5R), plays essential roles in the central nervous system (CNS) and ligands that activate dopamine receptors have been used to treat many neuronal disorders, yet no single structure of an agonist-bound dopamine receptor has been determined. Here we report the structures of human D3R in complex with an inhibitory G protein, either bound to a pan agonist, the Parkinson’s Disease drug pramipexole, or to a D3R-specific agonist, PD128907. The structures reveal distinct agonist binding modes from the antagonist-bound D3R structure and conformational signatures for ligand-induced receptor activation. Mutagenesis and homology modeling shed light into determinants of ligand specificity across dopamine receptors and the mechanism of selective G i protein coupling. Together with antagonist-bound structures, these results provide a template for the rational design of dopamine receptor-targeting ligands.
- Published
- 2021
33. Structures of the human dopamine D3 receptor-G
- Author
-
Peiyu, Xu, Sijie, Huang, Chunyou, Mao, Brian E, Krumm, X Edward, Zhou, Yangxia, Tan, Xi-Ping, Huang, Yongfeng, Liu, Dan-Dan, Shen, Yi, Jiang, Xuekui, Yu, Hualiang, Jiang, Karsten, Melcher, Bryan L, Roth, Xi, Cheng, Yan, Zhang, and H Eric, Xu
- Subjects
Models, Molecular ,Structure-Activity Relationship ,HEK293 Cells ,Pramipexole ,Protein Domains ,Multiprotein Complexes ,Cryoelectron Microscopy ,Oxazines ,Receptors, Dopamine D3 ,Humans ,Benzopyrans ,GTP-Binding Protein alpha Subunits, Gi-Go - Abstract
The dopamine system, including five dopamine receptors (D1R-D5R), plays essential roles in the central nervous system (CNS), and ligands that activate dopamine receptors have been used to treat many neuropsychiatric disorders. Here, we report two cryo-EM structures of human D3R in complex with an inhibitory G protein and bound to the D3R-selective agonists PD128907 and pramipexole, the latter of which is used to treat patients with Parkinson's disease. The structures reveal agonist binding modes distinct from the antagonist-bound D3R structure and conformational signatures for ligand-induced receptor activation. Mutagenesis and homology modeling illuminate determinants of ligand specificity across dopamine receptors and the mechanisms for G
- Published
- 2020
34. Structure of an AMPK complex in an inactive, ATP-bound state
- Author
-
Anthony A. Kossiakoff, Kaleeckal G. Harikumar, Jared Lamp, Laurence J. Miller, Abigail E. Ellis, Ryan D. Sheldon, Kelly Suino-Powell, Yan Yan, X. Edward Zhou, Joseph S. Brunzelle, Somnath Mukherjee, H. Eric Xu, Russell G. Jones, Scott J. Novick, Katarzyna Radziwon, Karsten Melcher, Patrick R. Griffin, Irving E. Vega, Ting-Hai Xu, and Timothy S. Strutzenberg
- Subjects
Adenosine monophosphate ,Models, Molecular ,Protein Conformation ,AMP-Activated Protein Kinases ,Protein Engineering ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,Immunoglobulin Fab Fragments ,0302 clinical medicine ,Adenosine Triphosphate ,Protein Domains ,Adenine nucleotide ,Humans ,Phosphorylation ,Protein kinase A ,030304 developmental biology ,0303 health sciences ,Multidisciplinary ,Kinase ,Cryoelectron Microscopy ,AMPK ,Adenosine Monophosphate ,Protein kinase domain ,chemistry ,Biophysics ,Adenosine triphosphate ,030217 neurology & neurosurgery - Abstract
How to catch a dynamic state AMP-activated protein kinase (AMPK) is a key sensor of energy status in eukaryotes. Its dynamic structure is regulated by allosteric factors including phosphorylation and binding of nucleotides and metabolites. Yan et al. developed conformation-specific antibodies that trap AMPK in a fully inactive state that has experienced a large, domain-level rotation. Biophysical experiments in cells and in vitro are consistent with the structural work and support a model in which the activation loop is fully exposed in the completely inactive, dephosphorylated state. These structures inform our understanding of the complex allosteric behavior in this crucial metabolic regulator. Science , abe7565, this issue p. 413
- Published
- 2020
35. Molecular mechanisms of fentanyl mediated β-arrestin biased signaling
- Author
-
Erli You, Parker W. de Waal, Karsten Melcher, H. Eric Xu, Jingjing Shi, Xiaoxi Wang, Bradley M. Dickson, and Yi Jiang
- Subjects
0301 basic medicine ,Receptors, Opioid, mu ,Molecular Dynamics ,Biochemistry ,Fentanyl ,Receptors, G-Protein-Coupled ,chemistry.chemical_compound ,0302 clinical medicine ,Computational Chemistry ,Cell Signaling ,Opioid receptor ,Biochemical Simulations ,Medicine and Health Sciences ,Biology (General) ,Amines ,Free Energy ,beta-Arrestins ,Analgesics ,Crystallography ,Ecology ,Chemistry ,Organic Compounds ,Physics ,Drugs ,Condensed Matter Physics ,Analgesics, Opioid ,DAMGO ,Computational Theory and Mathematics ,Modeling and Simulation ,Physical Sciences ,Crystal Structure ,Thermodynamics ,medicine.drug ,Research Article ,Signal Transduction ,Agonist ,Transmembrane Receptors ,G protein ,medicine.drug_class ,QH301-705.5 ,Computational biology ,Molecular Dynamics Simulation ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Structure-Activity Relationship ,GTP-Binding Proteins ,Genetics ,medicine ,Arrestin ,Solid State Physics ,Pain Management ,Humans ,Molecular Biology ,Ecology, Evolution, Behavior and Systematics ,G protein-coupled receptor ,Pharmacology ,Organic Chemistry ,Chemical Compounds ,Biology and Life Sciences ,Proteins ,Computational Biology ,Cell Biology ,G-Protein Signaling ,030104 developmental biology ,Structural biology ,Opioid ,G Protein Coupled Receptors ,Neuroscience ,030217 neurology & neurosurgery - Abstract
The development of novel analgesics with improved safety profiles to combat the opioid epidemic represents a central question to G protein coupled receptor structural biology and pharmacology: What chemical features dictate G protein or β-arrestin signaling? Here we use adaptively biased molecular dynamics simulations to determine how fentanyl, a potent β-arrestin biased agonist, binds the μ-opioid receptor (μOR). The resulting fentanyl-bound pose provides rational insight into a wealth of historical structure-activity-relationship on its chemical scaffold. Following an in-silico derived hypothesis we found that fentanyl and the synthetic opioid peptide DAMGO require M153 to induce β-arrestin coupling, while M153 was dispensable for G protein coupling. We propose and validate an activation mechanism where the n-aniline ring of fentanyl mediates μOR β-arrestin through a novel M153 “microswitch” by synthesizing fentanyl-based derivatives that exhibit complete, clinically desirable, G protein biased coupling. Together, these results provide molecular insight into fentanyl mediated β-arrestin biased signaling and a rational framework for further optimization of fentanyl-based analgesics with improved safety profiles., Author summary The global opioid crisis has drawn significant attention to the risks associated with over-use of synthetic opioids. Despite the public attention, and perhaps in-line with the profit-based incentives of the pharmaceutical industry, there is no public structure of mu-opioid receptor bound to fentanyl or fentanyl derivatives. A publicly available structure of the complex would allow open-source development of safer painkillers and synthetic antagonists. Current overdose antidotes, antagonists, require natural products in their synthesis which persists a sizable barrier to market and develop better antidotes. In this work we use advance molecular dynamics techniques to obtain the bound geometry of mu-opioid receptor with fentanyl (and derivatives) and uncovered a novel molecular switch involved in receptor activation. Based on our in-silico structure, we synthesized and tested novel compounds to validate our predicted structure. Herein we report the bound state of several dangerous fentanyl derivatives and introduce new derivatives with signaling profiles that may lead to lower risk of respiratory depression.
- Published
- 2020
36. Cryo-EM Structure of Human Cannabinoid Receptor CB2-Gi Signaling Complex
- Author
-
Heng Liu, Cheng Zhang, Xiang-Qun Xie, Terence F. McGuire, X. Edward Zhou, Karsten Melcher, Ying Xue, Changrui Xing, H. Eric Xu, Xing Meng, Maozi Chen, Ting-Hai Xu, Lei Wang, Gongpu Zhao, Junmei Wang, Youwen Zhuang, and Zhiwei Feng
- Subjects
Drug ,Agonist ,Cannabinoid receptor ,Cryo-electron microscopy ,medicine.drug_class ,media_common.quotation_subject ,Inflammation ,CHO Cells ,GTP-Binding Protein alpha Subunits, Gi-Go ,Spodoptera ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Article ,Receptor, Cannabinoid, CB2 ,03 medical and health sciences ,Cricetulus ,0302 clinical medicine ,Cricetinae ,Sf9 Cells ,medicine ,Animals ,Humans ,Cannabinoid Receptor Antagonists ,030304 developmental biology ,media_common ,Cannabinoid Receptor Agonists ,0303 health sciences ,Binding Sites ,Cryoelectron Microscopy ,Endocannabinoid system ,Molecular Docking Simulation ,Membrane protein ,Docking (molecular) ,lipids (amino acids, peptides, and proteins) ,medicine.symptom ,Neuroscience ,030217 neurology & neurosurgery ,Protein Binding ,Signal Transduction - Abstract
Drugs selectively targeting CB2 hold promise for treating neurodegenerative disorders, inflammation, and pain while avoiding psychotropic side effects mediated by CB1. The mechanisms underlying CB2 activation and signaling are poorly understood but critical for drug design. Here we report the cryo-EM structure of the human CB2-Gi signaling complex bound to the agonist WIN 55,212-2. The 3D structure reveals the binding mode of WIN 55,212-2 and structural determinants for distinguishing CB2 agonists from antagonists, which are supported by a pair of rationally designed agonist and antagonist. Further structural analyses with computational docking results uncover the differences between CB2 and CB1 in receptor activation, ligand recognition, and Gi coupling. These findings are expected to facilitate rational structure-based discovery of drugs targeting the cannabinoid system.
- Published
- 2020
37. Crystal structure of the Frizzled 4 receptor in a ligand-free state
- Author
-
H. Eric Xu, Lu Yang, Petr Popov, Raymond C. Stevens, Fei Xu, Mengchen Pu, Yuanzheng He, Yuxiang Chen, Kelly Suino-Powell, Shaowei Dong, Kaleeckal G. Harikumar, Yiran Wu, Parker W. de Waal, Laurence J. Miller, Zachary J. DeBruine, Yu Guo, Karsten Melcher, Vsevolod Katritch, Wenqing Shui, Gye Won Han, Shifan Yang, Saheem A. Zaidi, Suwen Zhao, Bingjie Zhang, and Ting-Hai Xu
- Subjects
Models, Molecular ,0301 basic medicine ,Frizzled ,Binding Sites ,Multidisciplinary ,Chemistry ,Protein domain ,Dishevelled Proteins ,Wnt signaling pathway ,Molecular Dynamics Simulation ,Crystallography, X-Ray ,Ligands ,Ligand (biochemistry) ,Frizzled Receptors ,Transmembrane protein ,03 medical and health sciences ,Transmembrane domain ,030104 developmental biology ,Protein Domains ,Biophysics ,Humans ,Cysteine ,Binding site ,Wnt Signaling Pathway ,G protein-coupled receptor - Abstract
Frizzled receptors (FZDs) are class-F G-protein-coupled receptors (GPCRs) that function in Wnt signalling and are essential for developing and adult organisms1,2. As central mediators in this complex signalling pathway, FZDs serve as gatekeeping proteins both for drug intervention and for the development of probes in basic and in therapeutic research. Here we present an atomic-resolution structure of the human Frizzled 4 receptor (FZD4) transmembrane domain in the absence of a bound ligand. The structure reveals an unusual transmembrane architecture in which helix VI is short and tightly packed, and is distinct from all other GPCR structures reported so far. Within this unique transmembrane fold is an extremely narrow and highly hydrophilic pocket that is not amenable to the binding of traditional GPCR ligands. We show that such a pocket is conserved across all FZDs, which may explain the long-standing difficulties in the development of ligands for these receptors. Molecular dynamics simulations on the microsecond timescale and mutational analysis uncovered two coupled, dynamic kinks located at helix VII that are involved in FZD4 activation. The stability of the structure in its ligand-free form, an unfavourable pocket for ligand binding and the two unusual kinks on helix VII suggest that FZDs may have evolved a novel ligand-recognition and activation mechanism that is distinct from that of other GPCRs.
- Published
- 2018
38. Crystal structure of the human 5-HT1B serotonin receptor bound to an inverse agonist
- Author
-
Bo Liu, Karsten Melcher, Xiaoxi Wang, Yechun Xu, Jiang Wang, Antao Dai, Ping Liu, Parker W. de Waal, Ming-Wei Wang, Xiaoqing Cai, Yanting Yin, X. Edward Zhou, Yu Zhou, H. Eric Xu, Wanchao Yin, Yi Jiang, Dehua Yang, Hong Liu, Martin Caffrey, Chia-Ying Huang, and Meitian Wang
- Subjects
0301 basic medicine ,Agonist ,medicine.drug_class ,Chemistry ,lcsh:Cytology ,Cell Biology ,Biochemistry ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Membrane protein ,Genetics ,medicine ,Extracellular ,Biophysics ,Inverse agonist ,Serotonin ,lcsh:QH573-671 ,Receptor ,Molecular Biology ,030217 neurology & neurosurgery ,5-HT receptor ,G protein-coupled receptor - Abstract
5-hydroxytryptamine (5-HT, also known as serotonin) regulates many physiological processes through the 5-HT receptor family. Here we report the crystal structure of 5-HT1B subtype receptor (5-HT1BR) bound to the psychotropic serotonin receptor inverse agonist methiothepin (MT). Crystallization was facilitated by replacing ICL3 with a novel optimized variant of BRIL (OB1) that enhances the formation of intermolecular polar interactions, making OB1 a potential useful tool for structural studies of membrane proteins. Unlike the agonist ergotamine (ERG), MT occupies only the conserved orthosteric binding pocket, explaining the wide spectrum effect of MT on serotonin receptors. Compared with ERG, MT shifts toward TM6 and sterically pushes residues W3276.48, F3306.50 and F3316.51 from inside the orthosteric binding pocket, leading to an outward movement of the extracellular end and a corresponding inward shift of the intracellular end of TM6, a feature shared by other reported inactive G protein-coupled receptor (GPCR) structures. Together with the previous agonist-bound serotonin receptor structures, the inverse agonist-bound 5-HT1BR structure identifies a basis for the ligand-mediated switch of 5-HT1BR activity and provides a structural understanding of the inactivation mechanism of 5-HT1BR and some other class A GPCRs, characterized by ligand-induced outward movement of the extracellular end of TM6 that is coupled with inward movement of the cytoplasmic end of this helix.
- Published
- 2018
39. Assembly and architecture of the Wnt/β-catenin signalosome at the membrane
- Author
-
Huaqiang Eric Xu, Karsten Melcher, and Zachary J. DeBruine
- Subjects
0301 basic medicine ,Pharmacology ,chemistry.chemical_classification ,Wnt signaling pathway ,Wnt signalling ,LRP5 ,Cell biology ,Dishevelled ,03 medical and health sciences ,030104 developmental biology ,Signalling ,chemistry ,Catenin ,Receptor ,Intracellular - Abstract
Wnt/β-catenin signalling is initiated by a ternary Wnt-Frizzled (FZD)-LDL receptor-related protein (LRP) 5/6 binding event. The resulting conformational changes in the FZD and LRP5/6 receptors promote the assembly of an intracellular signalosome driven by Dishevelled and Axin co-polymerization. Recent evidence suggests that the FZD receptor and LRP5/6 participate in the assembly of this signalosome by forming regulatory scaffolds for stabilizing Dishevelled and Axin adapters. In this review, we focus on the contributions of Wnts and their receptors in the assembly of the signalosome. We present an emerging model, which unifies Wnt receptor oligomerization with intracellular signalosome formation, and then discuss how FZD receptors might be targeted to either disrupt or enhance their capacity as a dynamic sensor of Wnt binding. Linked Articles This article is part of a themed section on WNT Signalling: Mechanisms and Therapeutic Opportunities. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.24/issuetoc
- Published
- 2017
40. Understanding the GPCR biased signaling through G protein and arrestin complex structures
- Author
-
Karsten Melcher, H. Eric Xu, and X. Edward Zhou
- Subjects
0301 basic medicine ,Arrestin ,Drug discovery ,G protein ,Biology ,Receptors, G-Protein-Coupled ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,GTP-Binding Proteins ,Structural Biology ,Cell surface receptor ,Molecular mechanism ,Animals ,Humans ,Signal transduction ,Receptor ,Molecular Biology ,hormones, hormone substitutes, and hormone antagonists ,Signal Transduction ,G protein-coupled receptor - Abstract
G protein-coupled receptors (GPCRs) are the largest family of cell surface receptors and are important drug targets for many human diseases. The determination of the 3-D structure of GPCRs and their signaling complexes has promoted our understanding of GPCR biology and provided templates for structure-based drug discovery. In this review, we focus on the recent structure work on GPCR signaling complexes, the β2-adrenoreceptor-Gs and the rhodopsin-arrestin complexes in particular, and highlight the structural features of GPCR complexes involved in G protein- and arrestin-mediated signal transduction. The crystal structures reveal distinct structural mechanisms by which GPCRs recruit a G protein and an arrestin. A comparison of the two complex structures provides insight into the molecular mechanism of functionally selective GPCR signaling, and a structural basis for the discovery of G protein- and arrestin-biased treatments of human diseases related to GPCR signal transduction.
- Published
- 2017
41. Rearrangement of a polar core provides a conserved mechanism for constitutive activation of class B G protein-coupled receptors
- Author
-
Karsten Melcher, Lihua Zhao, Parker W. de Waal, Xiaoqing Cai, Yuanzheng He, Ming-Wei Wang, Dehua Yang, H. Eric Xu, Yi Jiang, and Yanting Yin
- Subjects
Models, Molecular ,0301 basic medicine ,Cell signaling ,Protein Conformation ,Receptors, Vasoactive Intestinal Polypeptide, Type I ,G protein ,Stereochemistry ,Recombinant Fusion Proteins ,Ligands ,Receptors, Corticotropin-Releasing Hormone ,Second Messenger Systems ,Biochemistry ,Cell Line ,03 medical and health sciences ,Receptors, Glucagon ,Arrestin ,Humans ,Protein Interaction Domains and Motifs ,Editors' Picks ,Amino Acid Sequence ,Receptor ,Molecular Biology ,Conserved Sequence ,Receptor, Parathyroid Hormone, Type 1 ,G protein-coupled receptor ,Binding Sites ,030102 biochemistry & molecular biology ,biology ,Protein Stability ,Cell Biology ,Peptide Fragments ,Recombinant Proteins ,Transmembrane domain ,030104 developmental biology ,Amino Acid Substitution ,Structural Homology, Protein ,Rhodopsin ,Mutation ,Mutagenesis, Site-Directed ,biology.protein ,Biophysics ,Hydrophobic and Hydrophilic Interactions ,Glucagon receptor ,Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide, Type I - Abstract
The glucagon receptor (GCGR) belongs to the secretin-like (class B) family of G protein-coupled receptors (GPCRs) and is activated by the peptide hormone glucagon. The structures of an activated class B GPCR have remained unsolved, preventing a mechanistic understanding of how these receptors are activated. Using a combination of structural modeling and mutagenesis studies, we present here two modes of ligand-independent activation of GCGR. First, we identified a GCGR-specific hydrophobic lock comprising Met-338 and Phe-345 within the IC3 loop and transmembrane helix 6 (TM6) and found that this lock stabilizes the TM6 helix in the inactive conformation. Disruption of this hydrophobic lock led to constitutive G protein and arrestin signaling. Second, we discovered a polar core comprising conserved residues in TM2, TM3, TM6, and TM7, and mutations that disrupt this polar core led to constitutive GCGR activity. On the basis of these results, we propose a mechanistic model of GCGR activation in which TM6 is held in an inactive conformation by the conserved polar core and the hydrophobic lock. Mutations that disrupt these inhibitory elements allow TM6 to swing outward to adopt an active TM6 conformation similar to that of the canonical β2-adrenergic receptor complexed with G protein and to that of rhodopsin complexed with arrestin. Importantly, mutations in the corresponding polar core of several other members of class B GPCRs, including PTH1R, PAC1R, VIP1R, and CRFR1, also induce constitutive G protein signaling, suggesting that the rearrangement of the polar core is a conserved mechanism for class B GPCR activation.
- Published
- 2017
42. Molecular assembly of rhodopsin with G protein-coupled receptor kinases
- Author
-
Karsten Melcher, Kuntal Pal, Devrishi Goswami, Gongpu Zhao, H. Eric Xu, Yanting Yin, Xiang Gao, Li Hou, Oliver P. Ernst, Patrick R. Griffin, and Yuanzheng He
- Subjects
0301 basic medicine ,Rhodopsin ,Blotting, Western ,Mutant ,GRK1 ,Biology ,GRK5 ,Mass Spectrometry ,03 medical and health sciences ,GPCR ,Humans ,Binding site ,Molecular Biology ,G protein-coupled receptor ,G protein-coupled receptor kinase ,Binding Sites ,030102 biochemistry & molecular biology ,Kinase ,Cell Biology ,G-Protein-Coupled Receptor Kinases ,Cell biology ,Q41L ,HEK293 Cells ,030104 developmental biology ,Biochemistry ,Protein kinase domain ,biology.protein ,Phosphorylation ,Original Article ,Protein Binding - Abstract
G protein-coupled receptor kinases (GRKs) play pivotal roles in desensitizing GPCR signaling but little is known about how GRKs recognize and phosphorylate GPCRs due to the technical difficulties in detecting the highly dynamic GPCR/GRK interaction. By combining a genetic approach with multiple biochemical assays, we identified the key determinants for the assembly of the prototypical GPCR rhodopsin with its kinase GRK1. Our work reveals that the regulatory G-protein signaling homology (RH) domain of GRKs is the primary binding site to GPCRs and an active conformation of the GRK1 kinase domain is required for efficient interaction with rhodopsin. In addition, we provide a mechanistic solution for the longstanding puzzle about the gain-of-function Q41L mutation in GRK5. This mutation is in the RH domain and increases the capacity of the GRK mutant to interact with and to desensitize GPCRs. Finally we present the principal architecture of a rhodopsin/GRK complex through negative stain electron microscopy reconstruction. Together, these data define the key components for the rhodopsin/GRK1 interaction and provide a framework for understanding GRK-mediated desensitization of GPCRs.
- Published
- 2017
43. Wnt5a promotes Frizzled-4 signalosome assembly by stabilizing cysteine-rich domain dimerization
- Author
-
Jiyuan Ke, H. Eric Xu, Kaleeckal G. Harikumar, Laurence J. Miller, Bart O. Williams, Peter Borowsky, Karsten Melcher, Zachary J. DeBruine, Wenqing Xu, and Xin Gu
- Subjects
Models, Molecular ,0301 basic medicine ,Frizzled ,Biology ,Crystallography, X-Ray ,Wnt-5a Protein ,Fatty Acids, Monounsaturated ,Cell membrane ,Wnt signalosome assembly ,03 medical and health sciences ,Genetics ,medicine ,Humans ,Cysteine ,Receptor ,beta Catenin ,chemistry.chemical_classification ,Wnt signaling pathway ,LRP6 ,LRP5 ,Frizzled Receptors ,eye diseases ,Protein Structure, Tertiary ,Dishevelled ,Cell biology ,Wnt Proteins ,HEK293 Cells ,030104 developmental biology ,medicine.anatomical_structure ,chemistry ,Protein Multimerization ,Protein Binding ,Signal Transduction ,Research Paper ,Developmental Biology - Abstract
Wnt/β-catenin signaling is activated when extracellular Wnt ligands bind Frizzled (FZD) receptors at the cell membrane. Wnts bind FZD cysteine-rich domains (CRDs) with high affinity through a palmitoylated N-terminal “thumb” and a disulfide-stabilized C-terminal “index finger,” yet how these binding events trigger receptor activation and intracellular signaling remains unclear. Here we report the crystal structure of the Frizzled-4 (FZD4) CRD in complex with palmitoleic acid, which reveals a CRD tetramer consisting of two cross-braced CRD dimers. Each dimer is stabilized by interactions of one hydrophobic palmitoleic acid tail with two CRD palmitoleoyl-binding grooves oriented end to end, suggesting that the Wnt palmitoleoyl group stimulates CRD–CRD interaction. Using bioluminescence resonance energy transfer (BRET) in live cells, we show that WNT5A stimulates dimerization of membrane-anchored FZD4 CRDs and oligomerization of full-length FZD4, which requires the integrity of CRD palmitoleoyl-binding residues. These results suggest that FZD receptors may form signalosomes in response to Wnt binding through the CRDs and that the Wnt palmitoleoyl group is important in promoting these interactions. These results complement our understanding of lipoprotein receptor-related proteins 5 and 6 (LRP5/6), Dishevelled, and Axin signalosome assembly and provide a more complete model for Wnt signalosome assembly both intracellularly and at the membrane.
- Published
- 2017
44. Defining the minimum substrate and charge recognition model of gamma-secretase
- Author
-
H. Eric Xu, Ting-Hai Xu, Yan Yan, and Karsten Melcher
- Subjects
Phosphatidylinositol 4,5-Diphosphate ,0301 basic medicine ,Arginine ,amyloid precursor protein (APP) ,γ-secretase ,Cleavage (embryo) ,Substrate Specificity ,Amyloid beta-Protein Precursor ,03 medical and health sciences ,chemistry.chemical_compound ,Notch protein ,0302 clinical medicine ,Alzheimer Disease ,Amyloid precursor protein ,Humans ,Pharmacology (medical) ,Amino Acid Sequence ,Phosphatidylinositol ,Peptide sequence ,C99 ,Cells, Cultured ,Gamma secretase ,Pharmacology ,recognition model ,Amyloid beta-Peptides ,Receptors, Notch ,biology ,General Medicine ,Alzheimer's disease ,Peptide Fragments ,Transmembrane domain ,030104 developmental biology ,chemistry ,Biochemistry ,amyloid beta (Aβ) ,Biophysics ,biology.protein ,minimum substrate ,Original Article ,Amyloid Precursor Protein Secretases ,Amyloid precursor protein secretase ,030217 neurology & neurosurgery - Abstract
γ-Secretase is an intramembrane aspartyl protease that cleaves the C99 fragment of amyloid precursor protein to generate extracellular Aβ peptides. These peptides can oligomerize and aggregate to form amyloid plaques, processes that are widely believed to be causal for Alzheimer's disease. In spite of this critical function, it remains unknown how γ-secretase recognizes C99 and its other substrates, including Notch. In this study we determined E22-K55 as the minimal C99 fragment that was sufficient and required for initial cleavage. Within this fragment, we identified four determinants: (i) a transferable extracellular determinant that differed between C99 and Notch, and which included negative charge in the case of C99, (ii) the amino acid sequence of the C-terminal half of the transmembrane helix, (iii) an invariant lysine or arginine at the intracellular membrane border, and (iv) a positive charge cluster that included the invariant lysine/arginine. We demonstrated that the charge clusters of C99 and Notch receptors could directly bind phosphatidylinositol 4,5-bisphosphate (PIP2). The PIP2-binding cluster was required for γ-secretase cleavage, and modulation of membrane PIP2 levels strongly affected γ-secretase cleavage levels and the Aβ40/Aβ42 ratio, providing support for the importance of the PIP2 interaction in cells. Together, these studies provide critically needed insight into substrate recognition by γ-secretase.
- Published
- 2017
45. Structure of the PRC2 complex and application to drug discovery
- Author
-
Xiaoxi Wang, Yi Shi, H. Eric Xu, Youwen Zhuang, Karsten Melcher, and Yi Jiang
- Subjects
0301 basic medicine ,Antineoplastic Agents ,Peptide ,Review ,macromolecular substances ,Computational biology ,medicine.disease_cause ,Small Molecule Libraries ,03 medical and health sciences ,Histone H3 ,Neoplasms ,Drug Discovery ,medicine ,Humans ,Gene silencing ,Pharmacology (medical) ,Pharmacology ,chemistry.chemical_classification ,Genetics ,Mutation ,biology ,Drug discovery ,fungi ,Polycomb Repressive Complex 2 ,General Medicine ,Small molecule ,Chromatin ,030104 developmental biology ,chemistry ,biology.protein ,PRC2 - Abstract
The polycomb repressive complexes 2 (PRC2) complex catalyzes tri-methylation of histone H3 lysine 27 (H3K27), a repressive chromatin marker associated with gene silencing. Overexpression and mutations of PRC2 are found in a wide variety of cancers, making the catalytic activity of PRC2 an important target of cancer therapy. This review highlights recent structural breakthroughs of the human PRC2 complex bound to the H3K27 peptide and a small molecule inhibitor, which provide critically needed insight into PRC2-targeted drug discovery.
- Published
- 2017
46. Structural insights into alternative splicing-mediated desensitization of jasmonate signaling
- Author
-
H. Eric Xu, Sheng Yang He, Koichi Sugimoto, Mingguo Zhou, Feng Zhang, Li Zhang, Rongzhi Chen, Jiyuan Ke, Karsten Melcher, and Gregg A. Howe
- Subjects
Models, Molecular ,0106 biological sciences ,0301 basic medicine ,Protein Conformation ,Protein subunit ,Arabidopsis ,Repressor ,Cyclopentanes ,Crystallography, X-Ray ,Bioinformatics ,01 natural sciences ,03 medical and health sciences ,Ubiquitin ,Gene Expression Regulation, Plant ,Two-Hybrid System Techniques ,Amino Acid Sequence ,Oxylipins ,Jasmonate ,Isoleucine ,Transcription factor ,Derepression ,Multidisciplinary ,Sequence Homology, Amino Acid ,biology ,Arabidopsis Proteins ,Alternative splicing ,food and beverages ,Nuclear Proteins ,Biological Sciences ,Ubiquitin ligase ,Cell biology ,Repressor Proteins ,Alternative Splicing ,030104 developmental biology ,Trans-Activators ,biology.protein ,Protein Binding ,Signal Transduction ,010606 plant biology & botany - Abstract
Jasmonate ZIM-domain (JAZ) transcriptional repressors play a key role in regulating jasmonate (JA) signaling in plants. Below a threshold concentration of jasmonoyl isoleucine (JA-Ile), the active form of JA, the C-terminal Jas motif of JAZ proteins binds MYC transcription factors to repress JA signaling. With increasing JA-Ile concentration, the Jas motif binds to JA-Ile and the COI1 subunit of the SCFCOI1 E3 ligase, which mediates ubiquitination and proteasomal degradation of JAZ repressors, resulting in derepression of MYC transcription factors. JA signaling subsequently becomes desensitized, in part by feedback induction of JAZ splice variants that lack the C-terminal Jas motif but include an N-terminal cryptic MYC-interaction domain (CMID). The CMID sequence is dissimilar to the Jas motif and is incapable of recruiting SCFCOI1, allowing CMID-containing JAZ splice variants to accumulate in the presence of JA and to re-repress MYC transcription factors as an integral part of reestablishing signal homeostasis. The mechanism by which the CMID represses MYC transcription factors remains elusive. Here we describe the crystal structure of the MYC3-CMIDJAZ10 complex. In contrast to the Jas motif, which forms a single continuous helix when bound to MYC3, the CMID adopts a loop-helix-loop-helix architecture with modular interactions with both the Jas-binding groove and the backside of the Jas-interaction domain of MYC3. This clamp-like interaction allows the CMID to bind MYC3 tightly and block access of MED25 (a subunit of the Mediator coactivator complex) to the MYC3 transcriptional activation domain, shedding light on the enigmatic mechanism by which JAZ splice variants desensitize JA signaling.
- Published
- 2017
47. Crystallization of a Complex Between MYC and Jas Motif
- Author
-
Feng, Zhang, Sheng Yang, He, and Karsten, Melcher
- Subjects
Proto-Oncogene Proteins c-myc ,Repressor Proteins ,Basic Helix-Loop-Helix Leucine Zipper Transcription Factors ,Multiprotein Complexes ,Amino Acid Motifs ,Arabidopsis ,Gene Expression ,Amino Acid Sequence ,Cloning, Molecular ,Crystallization ,Recombinant Proteins - Abstract
In the jasmonate signaling pathway, a region of 17 amino acids within the Jas motif of JAZ proteins and a conserved region within the N-terminus of MYC proteins are sufficient for JAZ-MYC interactions. Crystal structures of Jas-MYC complexes have revealed the structural basis of this important interaction. Here, we describe methods of cloning, expression, and purification of MYC N-terminal proteins and their co-crystallization with Jas motif peptides.
- Published
- 2019
48. Crystallization of a Complex Between MYC and Jas Motif
- Author
-
Feng Zhang, Sheng Yang He, and Karsten Melcher
- Subjects
0106 biological sciences ,0301 basic medicine ,chemistry.chemical_classification ,Chemistry ,MYC Transcription Factor ,Myc proteins ,01 natural sciences ,Cell biology ,Amino acid ,Jasmonate signaling ,03 medical and health sciences ,030104 developmental biology ,Motif (music) ,Jasmonate ,010606 plant biology & botany - Abstract
In the jasmonate signaling pathway, a region of 17 amino acids within the Jas motif of JAZ proteins and a conserved region within the N-terminus of MYC proteins are sufficient for JAZ-MYC interactions. Crystal structures of Jas-MYC complexes have revealed the structural basis of this important interaction. Here, we describe methods of cloning, expression, and purification of MYC N-terminal proteins and their co-crystallization with Jas motif peptides.
- Published
- 2019
49. A complex structure of arrestin-2 bound to a G protein-coupled receptor
- Author
-
Mingliang Jin, Xiang Gao, P.W. de Waal, H. Eric Xu, Wanchao Yin, Jing Gao, Yanting Yin, Yulong Yin, Xiaoxi Wang, Yuanzheng He, Yan Zhang, Yao Cong, X. Edward Zhou, Zhihai Li, Kuntal Pal, Xuekui Yu, Hu Zhou, Yi Jiang, and Karsten Melcher
- Subjects
Neurotensin receptor 1 ,Protein Conformation ,Article ,03 medical and health sciences ,0302 clinical medicine ,Arrestin ,Humans ,Receptors, Neurotensin ,Homology modeling ,Receptor ,Molecular Biology ,030304 developmental biology ,G protein-coupled receptor ,0303 health sciences ,biology ,Chemistry ,Cell Biology ,beta-Arrestin 2 ,Molecular Docking Simulation ,Transmembrane domain ,Rhodopsin ,biology.protein ,ARR3 ,Biophysics ,030217 neurology & neurosurgery ,Protein Binding - Abstract
Arrestins comprise a family of signal regulators of G-protein-coupled receptors (GPCRs), which include arrestins 1 to 4. While arrestins 1 and 4 are visual arrestins dedicated to rhodopsin, arrestins 2 and 3 (Arr2 and Arr3) are β-arrestins known to regulate many nonvisual GPCRs. The dynamic and promiscuous coupling of Arr2 to nonvisual GPCRs has posed technical challenges to tackle the basis of arrestin binding to GPCRs. Here we report the structure of Arr2 in complex with neurotensin receptor 1 (NTSR1), which reveals an overall assembly that is strikingly different from the visual arrestin-rhodopsin complex by a 90° rotation of Arr2 relative to the receptor. In this new configuration, intracellular loop 3 (ICL3) and transmembrane helix 6 (TM6) of the receptor are oriented toward the N-terminal domain of the arrestin, making it possible for GPCRs that lack the C-terminal tail to couple Arr2 through their ICL3. Molecular dynamics simulation and crosslinking data further support the assembly of the Arr2–NTSR1 complex. Sequence analysis and homology modeling suggest that the Arr2–NTSR1 complex structure may provide an alternative template for modeling arrestin-GPCR interactions.
- Published
- 2019
- Full Text
- View/download PDF
50. Structural basis of Fusarium myosin I inhibition by phenamacril
- Author
-
Karsten Melcher, X. Edward Zhou, Yuanye Zhu, Joseph S. Brunzelle, Xiaoman Cao, H. Eric Xu, Feng Zhang, Gong Yuanping, Mingguo Zhou, and Yuxin Zhou
- Subjects
Adenosine Triphosphatase ,Fungal Structure ,Plant Science ,Pathology and Laboratory Medicine ,Biochemistry ,Contractile Proteins ,Fusarium ,Myosin ,Medicine and Health Sciences ,Cyanoacrylates ,Biology (General) ,Triticum ,Fungal Pathogens ,0303 health sciences ,Fungal protein ,Crystallography ,Physics ,030302 biochemistry & molecular biology ,Plant Fungal Pathogens ,food and beverages ,Condensed Matter Physics ,Enzymes ,Fungicide ,Medical Microbiology ,Physical Sciences ,Crystal Structure ,Pathogens ,Research Article ,QH301-705.5 ,Immunology ,Allosteric regulation ,Motor Proteins ,Actin Motors ,Plant Pathogens ,Fusarium Graminearum ,macromolecular substances ,Mycology ,Biology ,Myosins ,Zea mays ,Microbiology ,Fungal Proteins ,03 medical and health sciences ,Residue (chemistry) ,Myosin Type I ,Molecular Motors ,Virology ,Hydrolase ,Genetics ,Atpase activity ,Solid State Physics ,Molecular Biology ,Microbial Pathogens ,030304 developmental biology ,Plant Diseases ,Phosphatases ,Biology and Life Sciences ,Proteins ,Cell Biology ,RC581-607 ,Plant Pathology ,biology.organism_classification ,Fungicides, Industrial ,Cytoskeletal Proteins ,Enzymology ,Parasitology ,Immunologic diseases. Allergy - Abstract
Fusarium is a genus of filamentous fungi that includes species that cause devastating diseases in major staple crops, such as wheat, maize, rice, and barley, resulting in severe yield losses and mycotoxin contamination of infected grains. Phenamacril is a novel fungicide that is considered environmentally benign due to its exceptional specificity; it inhibits the ATPase activity of the sole class I myosin of only a subset of Fusarium species including the major plant pathogens F. graminearum, F. asiaticum and F. fujikuroi. To understand the underlying mechanisms of inhibition, species specificity, and resistance mutations, we have determined the crystal structure of phenamacril-bound F. graminearum myosin I. Phenamacril binds in the actin-binding cleft in a new allosteric pocket that contains the central residue of the regulatory Switch 2 loop and that is collapsed in the structure of a myosin with closed actin-binding cleft, suggesting that pocket occupancy blocks cleft closure. We have further identified a single, transferable phenamacril-binding residue found exclusively in phenamacril-sensitive myosins to confer phenamacril selectivity., Author summary Phenamacril is a recently identified myosin I inhibitor that is a potent and highly species-specific and myosin subtype-selective fungicide. We report the high-resolution structure of the phenamacril-bound myosin I motor domain of the major crop pathogen Fusarium graminearum, providing insight into the molecular mechanism of phenamacril action and resistance. These results are of broad significance for understanding the mode of actions of myosin-based fungicides and for designing novel myosin I inhibitors for crop protection and for treatment of human myosin dysfunction diseases.
- Published
- 2019
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.