251
|
Wytynck P, Rougé P, Van Damme EJM. Genome-wide screening of Oryza sativa ssp. japonica and indica reveals a complex family of proteins with ribosome-inactivating protein domains. PHYTOCHEMISTRY 2017; 143:87-97. [PMID: 28797946 DOI: 10.1016/j.phytochem.2017.07.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 07/08/2017] [Accepted: 07/22/2017] [Indexed: 06/07/2023]
Abstract
Ribosome-inactivating proteins (RIPs) are cytotoxic enzymes capable of halting protein synthesis by irreversible modification of ribosomes. Although RIPs are widespread they are not ubiquitous in the plant kingdom. The physiological importance of RIPs is not fully elucidated, but evidence suggests a role in the protection of the plant against biotic and abiotic stresses. Searches in the rice genome revealed a large and highly complex family of proteins with a RIP domain. A comparative analysis retrieved 38 RIP sequences from the genome sequence of Oryza sativa subspecies japonica and 34 sequences from the subspecies indica. The RIP sequences are scattered over different chromosomes but are mostly found on the third chromosome. The phylogenetic tree revealed the pairwise clustering of RIPs from japonica and indica. Molecular modeling and sequence analysis yielded information on the catalytic site of the enzyme, and suggested that a large part of RIP domains probably possess N-glycosidase activity. Several RIPs are differentially expressed in plant tissues and in response to specific abiotic stresses. This study provides an overview of RIP motifs in rice and will help to understand their biological role(s) and evolutionary relationships.
Collapse
Affiliation(s)
- Pieter Wytynck
- Lab Biochemistry and Glycobiology, Department of Molecular Biotechnology, Ghent University, Coupure Links 653, B-9000, Ghent, Belgium
| | - Pierre Rougé
- UMR152 PHARMA-DEV, Université de Toulouse, IRD, UPS, Chemin des Maraîchers 35, 31400, Toulouse, France
| | - Els J M Van Damme
- Lab Biochemistry and Glycobiology, Department of Molecular Biotechnology, Ghent University, Coupure Links 653, B-9000, Ghent, Belgium.
| |
Collapse
|
252
|
Tinklepaugh J, Smith BM, Hanlon E, Zubieta C, Bou-Abdallah F, Doyle RP. Exploring the Multiligand Binding Specificity of Saposin B Reveals Two Binding Sites. ACS OMEGA 2017; 2:7141-7145. [PMID: 29104953 PMCID: PMC5664142 DOI: 10.1021/acsomega.7b01334] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 10/11/2017] [Indexed: 06/07/2023]
Abstract
Saposin B (SapB) is a human lysosomal protein, critical for the degradation of O-sulfogalactosylceramide (sulfatide). SapB binds sulfatide and presents it to the active site of the enzyme arylsulfatase A. Deficiency of SapB leads to fatal activator-deficient metachromatic leukodystrophy. Given the conformational flexibility and the large hydrophobic "pocket" produced upon (physiologically relevant) homodimerization, SapB may have broader substrate diversity than originally thought. Herein, we present evidence using fluorescence spectroscopy and computational docking studies that SapB binds a wide variety of ligands at KD values varying from micromolar to nanomolar, with entropy being the primary driving force. We further demonstrate, for the first time, that SapB has two binding sites that can sequentially (and in a preferred order) accommodate up to two ligands at once.
Collapse
Affiliation(s)
- Jay Tinklepaugh
- Department
of Chemistry, Syracuse University, 111 College Place, Syracuse, New York 13244, United States
| | - Britannia M. Smith
- Department
of Chemistry, State University of New York
at Potsdam, Potsdam, New York 13676, United
States
| | - Etta Hanlon
- Department
of Chemistry, Syracuse University, 111 College Place, Syracuse, New York 13244, United States
| | - Chloe Zubieta
- Laboratoire
de Physiologie Cellulaire & Végétale, iRTSV, UMR 5168, CNRS/CEA/INRA/Univ. Grenoble Alpes, Grenoble 38054, France
| | - Fadi Bou-Abdallah
- Department
of Chemistry, State University of New York
at Potsdam, Potsdam, New York 13676, United
States
| | - Robert P. Doyle
- Department
of Chemistry, Syracuse University, 111 College Place, Syracuse, New York 13244, United States
- Department
of Medicine, State University of New York
at Upstate Medical University, Syracuse, New York 13210, United States
| |
Collapse
|
253
|
Wang N, Wang L, Xie XQ. ProSelection: A Novel Algorithm to Select Proper Protein Structure Subsets for in Silico Target Identification and Drug Discovery Research. J Chem Inf Model 2017; 57:2686-2698. [PMID: 29016123 DOI: 10.1021/acs.jcim.7b00277] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Molecular docking is widely applied to computer-aided drug design and has become relatively mature in the recent decades. Application of docking in modeling varies from single lead compound optimization to large-scale virtual screening. The performance of molecular docking is highly dependent on the protein structures selected. It is especially challenging for large-scale target prediction research when multiple structures are available for a single target. Therefore, we have established ProSelection, a docking preferred-protein selection algorithm, in order to generate the proper structure subset(s). By the ProSelection algorithm, protein structures of "weak selectors" are filtered out whereas structures of "strong selectors" are kept. Specifically, the structure which has a good statistical performance of distinguishing active ligands from inactive ligands is defined as a strong selector. In this study, 249 protein structures of 14 autophagy-related targets are investigated. Surflex-dock was used as the docking engine to distinguish active and inactive compounds against these protein structures. Both t test and Mann-Whitney U test were used to distinguish the strong from the weak selectors based on the normality of the docking score distribution. The suggested docking score threshold for active ligands (SDA) was generated for each strong selector structure according to the receiver operating characteristic (ROC) curve. The performance of ProSelection was further validated by predicting the potential off-targets of 43 U.S. Federal Drug Administration approved small molecule antineoplastic drugs. Overall, ProSelection will accelerate the computational work in protein structure selection and could be a useful tool for molecular docking, target prediction, and protein-chemical database establishment research.
Collapse
Affiliation(s)
- Nanyi Wang
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy; NIH National Center of Excellence for Computational Drug Abuse Research; Drug Discovery Institute, University of Pittsburgh , Pittsburgh, Pennsylvania 15260, United States
| | - Lirong Wang
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy; NIH National Center of Excellence for Computational Drug Abuse Research; Drug Discovery Institute, University of Pittsburgh , Pittsburgh, Pennsylvania 15260, United States
| | - Xiang-Qun Xie
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy; NIH National Center of Excellence for Computational Drug Abuse Research; Drug Discovery Institute, University of Pittsburgh , Pittsburgh, Pennsylvania 15260, United States
| |
Collapse
|
254
|
Götz KH, Hacker SM, Mayer D, Dürig JN, Stenger S, Marx A. Inhibitors of the Diadenosine Tetraphosphate Phosphorylase Rv2613c of Mycobacterium tuberculosis. ACS Chem Biol 2017; 12:2682-2689. [PMID: 28892605 DOI: 10.1021/acschembio.7b00653] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The intracellular concentration of diadenosine tetraphospate (Ap4A) increases upon exposure to stress conditions. Despite being discovered over 50 years ago, the cellular functions of Ap4A are still enigmatic. If and how the varied Ap4A is a signal and involved in the signaling pathways leading to an appropriate cellular response remain to be discovered. Because the turnover of Ap4A by Ap4A cleaving enzymes is rapid, small molecule inhibitors for these enzymes would provide tools for the more detailed study of the role of Ap4A. Here, we describe the development of a high-throughput screening assay based on a fluorogenic Ap4A substrate for the identification and optimization of small molecule inhibitors for Ap4A cleaving enzymes. As proof-of-concept we screened a library of over 42 000 compounds toward their inhibitory activity against the Ap4A phosphorylase (Rv2613c) of Mycobacterium tuberculosis (Mtb). A sulfanylacrylonitril derivative with an IC50 of 260 ± 50 nM in vitro was identified. Multiple derivatives were synthesized to further optimize their properties with respect to their in vitro IC50 values and their cytotoxicity against human cells (HeLa). In addition, we selected two hits to study their antimycobacterial activity against virulent Mtb to show that they might be candidates for further development of antimycobacterial agents against multidrug-resistant Mtb.
Collapse
Affiliation(s)
- Kathrin H. Götz
- Department
of Chemistry, Konstanz Research School Chemical Biology, University of Konstanz, Universitätsstrasse 10, D-78464 Konstanz, Germany
| | - Stephan M. Hacker
- Department
of Chemistry, Konstanz Research School Chemical Biology, University of Konstanz, Universitätsstrasse 10, D-78464 Konstanz, Germany
| | - Daniel Mayer
- Institute
for Medical Microbiology and Hygiene, University Hospital of Ulm, Albert-Einstein-Allee
11, D-89081 Ulm, Germany
| | - Jan-Niklas Dürig
- Department
of Chemistry, Konstanz Research School Chemical Biology, University of Konstanz, Universitätsstrasse 10, D-78464 Konstanz, Germany
| | - Steffen Stenger
- Institute
for Medical Microbiology and Hygiene, University Hospital of Ulm, Albert-Einstein-Allee
11, D-89081 Ulm, Germany
| | - Andreas Marx
- Department
of Chemistry, Konstanz Research School Chemical Biology, University of Konstanz, Universitätsstrasse 10, D-78464 Konstanz, Germany
| |
Collapse
|
255
|
Manalo RVM. Molecular interactions with redox sites and salt bridges modulate the anti-aggregatory effect of flavonoid, tannin and cardenolide moieties against amyloid-beta (1-42) in silico. In Silico Pharmacol 2017; 5:11. [PMID: 29085768 DOI: 10.1007/s40203-017-0033-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 10/09/2017] [Indexed: 12/14/2022] Open
Abstract
In this study, the interactions of flavonoid, tannin and cardenolide moieties as well as their known metabolites were docked against the apolar NMR structure of the aggregatory amyloid-beta fragment (Aβ1-42). Results showed that the catechin moiety favorably bound Aβ1-42 peptide at Asp23, Asn27, Ser26 and Glu22 residues, with chalcone similarly binding the middle region of the peptide. Remarkably, hippuric and ferulic acids exhibited hydrophobic interactions with Aβ1-42 at the latter portion of the peptide, possibly blocking the salt bridges formed by Glu22-Lys28 which stabilizes Phe19-Gly25, as well as the β-sheet Leu34-Gly38 that are known to exist in peptide aggregation. Meanwhile, the metabolites of hydrolyzable tannins, such as urolithin A and gallic acid, exhibited H-bonding interactions with different residues of Aβ1-42, including Asp1, Asp23 and hydrophobic interactions by gallic acid planar ring to the Hsd6 residue. The coverage was lessened in pyrogallol, suggesting that gallic acid loses its efficacy when further metabolized. Lastly, the different binding poses of the cardenolide moiety interacted with Hsp6 (protonated His) and Tyr10 via hydrophobic interactions. Due to these interactions, the large polycyclic moiety of the ligand would also block further interactions with Hsd6 (prototropic tautomer of His), Asp7, Ser8 and Gly9 that are integral to His6-His13-His14, Arg5-Asp7and Leu34-Gly38 β-sheets, salt bridges in Glu22-Lys28 and turn conformation Phe19-Gly25. Together, these data suggest that the known metabolites of anthocyanins and hydrolyzable tannins contribute the most effective anti-aggregatory interactions with Aβ1-42, with an unexpected role for cardiac glycosides such as the cardenolie moiety. These bring to light the important role of metabolism in vivo, and suggests further investigation on the effects of these metabolites when concentrated in vivo.
Collapse
Affiliation(s)
- Rafael Vincent M Manalo
- Department of Biochemistry and Molecular Biology, College of Medicine, University of the Philippines Manila, Ermita, 1000 Manila, Philippines
| |
Collapse
|
256
|
Fattahian Y, Riahi-Madvar A, Mirzaee R, Asadikaram G, Rahbar MR. In silico locating the immune-reactive segments of Lepidium draba peroxidase and designing a less immune-reactive enzyme derivative. Comput Biol Chem 2017; 70:21-30. [DOI: 10.1016/j.compbiolchem.2017.07.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2017] [Revised: 06/14/2017] [Accepted: 07/12/2017] [Indexed: 12/24/2022]
|
257
|
Biochemical Regulatory Features of Activation-Induced Cytidine Deaminase Remain Conserved from Lampreys to Humans. Mol Cell Biol 2017; 37:MCB.00077-17. [PMID: 28716949 DOI: 10.1128/mcb.00077-17] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 07/10/2017] [Indexed: 01/17/2023] Open
Abstract
Activation-induced cytidine deaminase (AID) is a genome-mutating enzyme that initiates class switch recombination and somatic hypermutation of antibodies in jawed vertebrates. We previously described the biochemical properties of human AID and found that it is an unusual enzyme in that it exhibits binding affinities for its substrate DNA and catalytic rates several orders of magnitude higher and lower, respectively, than a typical enzyme. Recently, we solved the functional structure of AID and demonstrated that these properties are due to nonspecific DNA binding on its surface, along with a catalytic pocket that predominantly assumes a closed conformation. Here we investigated the biochemical properties of AID from a sea lamprey, nurse shark, tetraodon, and coelacanth: representative species chosen because their lineages diverged at the earliest critical junctures in evolution of adaptive immunity. We found that these earliest-diverged AID orthologs are active cytidine deaminases that exhibit unique substrate specificities and thermosensitivities. Significant amino acid sequence divergence among these AID orthologs is predicted to manifest as notable structural differences. However, despite major differences in sequence specificities, thermosensitivities, and structural features, all orthologs share the unusually high DNA binding affinities and low catalytic rates. This absolute conservation is evidence for biological significance of these unique biochemical properties.
Collapse
|
258
|
Bhattacharya D, Svechkarev D, Souchek JJ, Hill TK, Taylor MA, Natarajan A, Mohs AM. Impact of structurally modifying hyaluronic acid on CD44 interaction. J Mater Chem B 2017; 5:8183-8192. [PMID: 29354263 PMCID: PMC5773055 DOI: 10.1039/c7tb01895a] [Citation(s) in RCA: 121] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
CD44 is a widely-distributed type I transmembrane glycoprotein that binds hyaluronic acid (HA) in most cell types, including primary tumor cells and cancer-initiating cells and has roles in cell migration, cell-cell, and cell-matrix adhesion. HA-derived conjugates and nanoparticles that target the CD44 receptor on cells have been reported for targeted delivery of therapeutics and imaging agents. Altering crucial interactions of HA with CD44 active sites holds significant importance in modulating targeting ability of hyaluronic acid to other cancer types that do not express the CD44 receptor or minimizing the interaction with CD44+ cells that are not target cells. The approach adopted here was deacetylation of the N-acetyl group and selective sulfation on the C6-OH on the HA polymer, which form critical interactions with the CD44 active site. Major interactions identified by molecular modeling were confirmed to be hydrogen bonding of the C6-OH with Tyr109 and hydrophobic interaction of the N-acetyl group with Tyr46, 83 and Ile 92. Modified HA was synthesized and characterized and its interactions were assessed by in vitro and molecular modeling approaches. In vitro techniques included flow cytometry and fluorescence polarization, while in silico approaches included docking and binding calculations by a MM-PBSA approach. These studies indicated that while both deacetylation and sulfation of HA individually decrease CD44 interaction, both chemical modifications are required to minimize interaction with CD44+ cells. The results of this study represent the first step to effective retargeting of HA-derived NPs for imaging and drug delivery.
Collapse
Affiliation(s)
- D. Bhattacharya
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198-6858, USA
| | - D. Svechkarev
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198-6858, USA
| | - J. J. Souchek
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198-6858, USA
| | - T. K. Hill
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198-6858, USA
| | - M. A. Taylor
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198-6858, USA
| | - A. Natarajan
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198-6858, USA
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198-6858, USA
| | - A. M. Mohs
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198-6858, USA
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198-6858, USA
- Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-6858, USA
| |
Collapse
|
259
|
Delannée V, Langouët S, Théret N, Siegel A. A modeling approach to evaluate the balance between bioactivation and detoxification of MeIQx in human hepatocytes. PeerJ 2017; 5:e3703. [PMID: 28879062 PMCID: PMC5582613 DOI: 10.7717/peerj.3703] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 07/27/2017] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Heterocyclic aromatic amines (HAA) are environmental and food contaminants that are potentially carcinogenic for humans. 2-Amino-3,8-dimethylimidazo[4,5-f]quinoxaline (MeIQx) is one of the most abundant HAA formed in cooked meat. MeIQx is metabolized by cytochrome P450 1A2 in the human liver into detoxificated and bioactivated products. Once bioactivated, MeIQx metabolites can lead to DNA adduct formation responsible for further genome instability. METHODS Using a computational approach, we developed a numerical model for MeIQx metabolism in the liver that predicts the MeIQx biotransformation into detoxification or bioactivation pathways according to the concentration of MeIQx. RESULTS Our results demonstrate that (1) the detoxification pathway predominates, (2) the ratio between detoxification and bioactivation pathways is not linear and shows a maximum at 10 µM of MeIQx in hepatocyte cell models, and (3) CYP1A2 is a key enzyme in the system that regulates the balance between bioactivation and detoxification. Our analysis suggests that such a ratio could be considered as an indicator of MeIQx genotoxicity at a low concentration of MeIQx. CONCLUSIONS Our model permits the investigation of the balance between bioactivation (i.e., DNA adduct formation pathway through the prediction of potential genotoxic compounds) and detoxification of MeIQx in order to predict the behaviour of this environmental contaminant in the human liver. It highlights the importance of complex regulations of enzyme competitions that should be taken into account in any further multi-organ models.
Collapse
Affiliation(s)
- Victorien Delannée
- UMR 6074 IRISA, CNRS, INRIA, University of Rennes 1, Rennes, France.,UMR Inserm U1085 IRSET, University of Rennes 1, Rennes, France
| | - Sophie Langouët
- UMR Inserm U1085 IRSET, University of Rennes 1, Rennes, France
| | - Nathalie Théret
- UMR 6074 IRISA, CNRS, INRIA, University of Rennes 1, Rennes, France.,UMR Inserm U1085 IRSET, University of Rennes 1, Rennes, France
| | - Anne Siegel
- UMR 6074 IRISA, CNRS, INRIA, University of Rennes 1, Rennes, France
| |
Collapse
|
260
|
Palmioli A, Ciaramelli C, Tisi R, Spinelli M, De Sanctis G, Sacco E, Airoldi C. Natural Compounds in Cancer Prevention: Effects of Coffee Extracts and Their Main Polyphenolic Component, 5-O-Caffeoylquinic Acid, on Oncogenic Ras Proteins. Chem Asian J 2017; 12:2457-2466. [PMID: 28719146 DOI: 10.1002/asia.201700844] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 07/17/2017] [Indexed: 01/08/2023]
Abstract
Recent epidemiological studies have demonstrated that the consumption of healthy foods that are particularly rich in polyphenols might reduce the incidence of cancer and neurodegenerative diseases. In particular, chlorogenic acids (CGAs) occur ubiquitously in food and represent the most abundant polyphenols in the human diet. A number of beneficial biological effects of CGAs, such as anti-inflammatory activity, anti-carcinogenic activity, and protection against neurodegenerative diseases, have been reported. However, the molecular mechanisms at the base of these biological activities have not yet been investigated in depth. By combining NMR spectroscopy, molecular docking, surface plasmon resonance and ex vivo assays of the Ras-dependent breast cancer cell line MDA-MB-231, we contribute to the elucidation of the molecular basis of the activity of CGAs and natural extracts from green and roasted coffee beans as chemoprotective dietary supplements.
Collapse
Affiliation(s)
- Alessandro Palmioli
- Department of Biotechnology and Biosciences, Università degli Studi di Milano-Bicocca, Piazza della Scienza 2, 20126, Milan, Italy
| | - Carlotta Ciaramelli
- Department of Biotechnology and Biosciences, Università degli Studi di Milano-Bicocca, Piazza della Scienza 2, 20126, Milan, Italy
| | - Renata Tisi
- Department of Biotechnology and Biosciences, Università degli Studi di Milano-Bicocca, Piazza della Scienza 2, 20126, Milan, Italy.,NeuroMI Milan Center for Neuroscience, University of Milano-Bicocca, 20126, Milano, Italy
| | - Michela Spinelli
- Department of Biotechnology and Biosciences, Università degli Studi di Milano-Bicocca, Piazza della Scienza 2, 20126, Milan, Italy
| | - Gaia De Sanctis
- Department of Biotechnology and Biosciences, Università degli Studi di Milano-Bicocca, Piazza della Scienza 2, 20126, Milan, Italy
| | - Elena Sacco
- Department of Biotechnology and Biosciences, Università degli Studi di Milano-Bicocca, Piazza della Scienza 2, 20126, Milan, Italy.,SysBio Center of Systems Biology, Piazza della Scienza 2, 20126, Milan, Italy
| | - Cristina Airoldi
- Department of Biotechnology and Biosciences, Università degli Studi di Milano-Bicocca, Piazza della Scienza 2, 20126, Milan, Italy.,NeuroMI Milan Center for Neuroscience, University of Milano-Bicocca, 20126, Milano, Italy.,SysBio Center of Systems Biology, Piazza della Scienza 2, 20126, Milan, Italy
| |
Collapse
|
261
|
Lin WK, Bolton EL, Cortopassi WA, Wang Y, O'Brien F, Maciejewska M, Jacobson MP, Garnham C, Ruas M, Parrington J, Lei M, Sitsapesan R, Galione A, Terrar DA. Synthesis of the Ca 2+-mobilizing messengers NAADP and cADPR by intracellular CD38 enzyme in the mouse heart: Role in β-adrenoceptor signaling. J Biol Chem 2017; 292:13243-13257. [PMID: 28539361 PMCID: PMC5555186 DOI: 10.1074/jbc.m117.789347] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 05/13/2017] [Indexed: 11/28/2022] Open
Abstract
Nicotinic acid adenine dinucleotide phosphate (NAADP) and cyclic ADP-ribose (cADPR) are Ca2+-mobilizing messengers important for modulating cardiac excitation-contraction coupling and pathophysiology. CD38, which belongs to the ADP-ribosyl cyclase family, catalyzes synthesis of both NAADP and cADPR in vitro However, it remains unclear whether this is the main enzyme for their production under physiological conditions. Here we show that membrane fractions from WT but not CD38-/- mouse hearts supported NAADP and cADPR synthesis. Membrane permeabilization of cardiac myocytes with saponin and/or Triton X-100 increased NAADP synthesis, indicating that intracellular CD38 contributes to NAADP production. The permeabilization also permitted immunostaining of CD38, with a striated pattern in WT myocytes, whereas CD38-/- myocytes and nonpermeabilized WT myocytes showed little or no staining, without striation. A component of β-adrenoreceptor signaling in the heart involves NAADP and lysosomes. Accordingly, in the presence of isoproterenol, Ca2+ transients and contraction amplitudes were smaller in CD38-/- myocytes than in the WT. In addition, suppressing lysosomal function with bafilomycin A1 reduced the isoproterenol-induced increase in Ca2+ transients in cardiac myocytes from WT but not CD38-/- mice. Whole hearts isolated from CD38-/- mice and exposed to isoproterenol showed reduced arrhythmias. SAN4825, an ADP-ribosyl cyclase inhibitor that reduces cADPR and NAADP synthesis in mouse membrane fractions, was shown to bind to CD38 in docking simulations and reduced the isoproterenol-induced arrhythmias in WT hearts. These observations support generation of NAADP and cADPR by intracellular CD38, which contributes to effects of β-adrenoreceptor stimulation to increase both Ca2+ transients and the tendency to disturb heart rhythm.
Collapse
Affiliation(s)
- Wee K Lin
- From the Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, United Kingdom
| | - Emma L Bolton
- From the Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, United Kingdom
| | - Wilian A Cortopassi
- the Department of Chemistry, Chemistry Research Laboratory, University of Oxford, Mansfield Road, Oxford OX1 3TA, United Kingdom
- the Department of Pharmaceutical Chemistry, University of California, San Francisco, California 94158, and
| | - Yanwen Wang
- the Faculty of Biology, Medicine, and Health, University of Manchester, Manchester M13 9NT, United Kingdom
| | - Fiona O'Brien
- From the Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, United Kingdom
| | - Matylda Maciejewska
- From the Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, United Kingdom
| | - Matthew P Jacobson
- the Department of Pharmaceutical Chemistry, University of California, San Francisco, California 94158, and
| | - Clive Garnham
- From the Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, United Kingdom
| | - Margarida Ruas
- From the Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, United Kingdom
| | - John Parrington
- From the Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, United Kingdom
| | - Ming Lei
- From the Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, United Kingdom
| | - Rebecca Sitsapesan
- From the Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, United Kingdom
| | - Antony Galione
- From the Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, United Kingdom
| | - Derek A Terrar
- From the Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, United Kingdom,
| |
Collapse
|
262
|
Structural basis of Notch O-glucosylation and O-xylosylation by mammalian protein-O-glucosyltransferase 1 (POGLUT1). Nat Commun 2017; 8:185. [PMID: 28775322 PMCID: PMC5543122 DOI: 10.1038/s41467-017-00255-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 06/14/2017] [Indexed: 11/08/2022] Open
Abstract
Protein O-glucosyltransferase 1/Rumi-mediated glucosylation of Notch epidermal growth factor-like (EGF-like) domains plays an important role in Notch signaling. Protein O-glucosyltransferase 1 shows specificity for folded EGF-like domains, it can only glycosylate serine residues in the C1XSXPC2 motif, and it possesses an uncommon dual donor substrate specificity. Using several EGF-like domains and donor substrate analogs, we have determined the structures of human Protein O-glucosyltransferase 1 substrate/product complexes that provide mechanistic insight into the basis for these properties. Notably, we show that Protein O-glucosyltransferase 1’s requirement for folded EGF-like domains also leads to its serine specificity and that two distinct local conformational states are likely responsible for its ability to transfer both glucose and xylose. We also show that Protein O-glucosyltransferase 1 possesses the potential to xylosylate a much broader range of EGF-like domain substrates than was previously thought. Finally, we show that Protein O-glucosyltransferase 1 has co-evolved with EGF-like domains of the type found in Notch. POGLUT1 is a protein-O-glucosyltransferase that transfers glucose and xylose to the EGF-like domains of Notch and other signaling receptors. Here the authors report the structure of human POGLUT1 in complexes with 3 different EGF-like domains and donor substrates and shed light on the enzyme’s substrate specificity and catalytic mechanism
Collapse
|
263
|
Ledbetter RN, Garcia Costas AM, Lubner CE, Mulder DW, Tokmina-Lukaszewska M, Artz JH, Patterson A, Magnuson TS, Jay ZJ, Duan HD, Miller J, Plunkett MH, Hoben JP, Barney BM, Carlson RP, Miller AF, Bothner B, King PW, Peters JW, Seefeldt LC. The Electron Bifurcating FixABCX Protein Complex from Azotobacter vinelandii: Generation of Low-Potential Reducing Equivalents for Nitrogenase Catalysis. Biochemistry 2017; 56:4177-4190. [PMID: 28704608 DOI: 10.1021/acs.biochem.7b00389] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The biological reduction of dinitrogen (N2) to ammonia (NH3) by nitrogenase is an energetically demanding reaction that requires low-potential electrons and ATP; however, pathways used to deliver the electrons from central metabolism to the reductants of nitrogenase, ferredoxin or flavodoxin, remain unknown for many diazotrophic microbes. The FixABCX protein complex has been proposed to reduce flavodoxin or ferredoxin using NADH as the electron donor in a process known as electron bifurcation. Herein, the FixABCX complex from Azotobacter vinelandii was purified and demonstrated to catalyze an electron bifurcation reaction: oxidation of NADH (Em = -320 mV) coupled to reduction of flavodoxin semiquinone (Em = -460 mV) and reduction of coenzyme Q (Em = 10 mV). Knocking out fix genes rendered Δrnf A. vinelandii cells unable to fix dinitrogen, confirming that the FixABCX system provides another route for delivery of electrons to nitrogenase. Characterization of the purified FixABCX complex revealed the presence of flavin and iron-sulfur cofactors confirmed by native mass spectrometry, electron paramagnetic resonance spectroscopy, and transient absorption spectroscopy. Transient absorption spectroscopy further established the presence of a short-lived flavin semiquinone radical, suggesting that a thermodynamically unstable flavin semiquinone may participate as an intermediate in the transfer of an electron to flavodoxin. A structural model of FixABCX, generated using chemical cross-linking in conjunction with homology modeling, revealed plausible electron transfer pathways to both high- and low-potential acceptors. Overall, this study informs a mechanism for electron bifurcation, offering insight into a unique method for delivery of low-potential electrons required for energy-intensive biochemical conversions.
Collapse
Affiliation(s)
- Rhesa N Ledbetter
- Department of Chemistry and Biochemistry, Utah State University , Logan, Utah 84322, United States
| | - Amaya M Garcia Costas
- Department of Chemistry and Biochemistry, Montana State University , Bozeman, Montana 59717, United States
| | - Carolyn E Lubner
- National Renewable Energy Laboratory , Golden, Colorado 80401, United States
| | - David W Mulder
- National Renewable Energy Laboratory , Golden, Colorado 80401, United States
| | - Monika Tokmina-Lukaszewska
- Department of Chemistry and Biochemistry, Montana State University , Bozeman, Montana 59717, United States
| | - Jacob H Artz
- Institute of Biological Chemistry, Washington State University , Pullman, Washington 99163, United States
| | - Angela Patterson
- Department of Chemistry and Biochemistry, Montana State University , Bozeman, Montana 59717, United States
| | - Timothy S Magnuson
- Department of Biological Sciences, Idaho State University , Pocatello, Idaho 83201, United States
| | - Zackary J Jay
- Department of Chemical and Biological Engineering, Center for Biofilm Engineering and Thermal Biology Institute, Montana State University , Bozeman, Montana 59717, United States
| | - H Diessel Duan
- Department of Chemistry, University of Kentucky , Lexington, Kentucky 40506, United States
| | - Jacquelyn Miller
- Department of Chemistry and Biochemistry, Montana State University , Bozeman, Montana 59717, United States
| | - Mary H Plunkett
- Department of Bioproducts and Biosystems Engineering and Biotechnology Institute, University of Minnesota , St. Paul, Minnesota 55108, United States
| | - John P Hoben
- Department of Chemistry, University of Kentucky , Lexington, Kentucky 40506, United States
| | - Brett M Barney
- Department of Bioproducts and Biosystems Engineering and Biotechnology Institute, University of Minnesota , St. Paul, Minnesota 55108, United States
| | - Ross P Carlson
- Department of Chemical and Biological Engineering, Center for Biofilm Engineering and Thermal Biology Institute, Montana State University , Bozeman, Montana 59717, United States
| | - Anne-Frances Miller
- Department of Chemistry, University of Kentucky , Lexington, Kentucky 40506, United States
| | - Brian Bothner
- Department of Chemistry and Biochemistry, Montana State University , Bozeman, Montana 59717, United States
| | - Paul W King
- National Renewable Energy Laboratory , Golden, Colorado 80401, United States
| | - John W Peters
- Department of Chemistry and Biochemistry, Montana State University , Bozeman, Montana 59717, United States.,Institute of Biological Chemistry, Washington State University , Pullman, Washington 99163, United States
| | - Lance C Seefeldt
- Department of Chemistry and Biochemistry, Utah State University , Logan, Utah 84322, United States
| |
Collapse
|
264
|
One-pot, four-component synthesis of spiroindoloquinazoline derivatives as phospholipase inhibitors. Tetrahedron 2017. [DOI: 10.1016/j.tet.2017.07.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
265
|
|
266
|
Liao X, Wang W, Fan C, Yang N, Zhao J, Zhang Y, Gao R, Shen G, Xia S, Li G. Prokaryotic expression, purification and characterization of human cyclooxygenase-2. Int J Mol Med 2017; 40:75-82. [PMID: 28560423 PMCID: PMC5466394 DOI: 10.3892/ijmm.2017.3007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 05/26/2017] [Indexed: 01/06/2023] Open
Abstract
Cyclooxygenase-2 (COX-2) is a key enzyme which catalyzes the conversion of arachidonic acid (AA) into prostaglandins (PGs). It plays an important role in pathophysiological processes, such as tumorigenesis, angiogenesis, inflammation and tumor cell drug resistance. Therefore, COX-2 has been viewed as an important target for cancer therapy. The preparation of COX-2 protein is an important initial step for the subsequent development of COX-2 inhibitors. In this study, we report a strategy to heterologously express truncated human COX-2 (trCOX-2) in Escherichia coli (E. coli) BL21(DE3) host cells. Following denaturation, purification and renaturation, we successfully obtained enzymatically active trCOX-2 containing 257 residues of the C-terminus. Homology modeling and molecular docking analyses revealed that trCOX-2 retained the predicted 3D catalytic domain structure and AA could still bind to its hydrophobic groove. Western blot analysis and ELISA indicated that the trCOX-2 still retained its characteristic antigenicity and binding activity, while COX assays revealed that trCOX-2 maintained its enzyme activity. On the whole, in this study, we provided a novel method to isolate trCOX-2 possessing AA binding and catalytic activities. This study thus lays a foundation to facilitate further investigations of COX-2 and offers a valuable method with which to achieve the prokaryotic expression of a eukaryotic membrane protein.
Collapse
Affiliation(s)
- Xiangzhi Liao
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, College of Life Sciences, Jilin University, Changchun, Jilin 130012, P.R. China
| | - Wenhan Wang
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, College of Life Sciences, Jilin University, Changchun, Jilin 130012, P.R. China
| | - Chuanxi Fan
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, College of Life Sciences, Jilin University, Changchun, Jilin 130012, P.R. China
| | - Ning Yang
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, College of Life Sciences, Jilin University, Changchun, Jilin 130012, P.R. China
| | - Jialiang Zhao
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, College of Life Sciences, Jilin University, Changchun, Jilin 130012, P.R. China
| | - Ying Zhang
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, College of Life Sciences, Jilin University, Changchun, Jilin 130012, P.R. China
| | - Ruijuan Gao
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, College of Life Sciences, Jilin University, Changchun, Jilin 130012, P.R. China
| | - Guannan Shen
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, College of Life Sciences, Jilin University, Changchun, Jilin 130012, P.R. China
| | - Simin Xia
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, College of Life Sciences, Jilin University, Changchun, Jilin 130012, P.R. China
| | - Guiying Li
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, College of Life Sciences, Jilin University, Changchun, Jilin 130012, P.R. China
| |
Collapse
|
267
|
Characterization of the Zika virus two-component NS2B-NS3 protease and structure-assisted identification of allosteric small-molecule antagonists. Antiviral Res 2017; 143:218-229. [PMID: 28461069 DOI: 10.1016/j.antiviral.2017.04.015] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 04/20/2017] [Accepted: 04/26/2017] [Indexed: 11/22/2022]
Abstract
The recent re-emergence of Zika virus (ZIKV)1, a member of the Flaviviridae family, has become a global emergency. Currently, there are no effective methods of preventing or treating ZIKV infection, which causes severe neuroimmunopathology and is particularly harmful to the developing fetuses of infected pregnant women. However, the pathology induced by ZIKV is unique among flaviviruses, and knowledge of the biology of other family members cannot easily be extrapolated to ZIKV. Thus, structure-function studies of ZIKV proteins are urgently needed to facilitate the development of effective preventative and therapeutic agents. Like other flaviviruses, ZIKV expresses an NS2B-NS3 protease, which consists of the NS2B cofactor and the NS3 protease domain and is essential for cleavage of the ZIKV polyprotein precursor and generation of fully functional viral proteins. Here, we report the enzymatic characterization of ZIKV protease, and we identify structural scaffolds for allosteric small-molecule inhibitors of this protease. Molecular modeling of the protease-inhibitor complexes suggests that these compounds bind to the druggable cavity in the NS2B-NS3 protease interface and affect productive interactions of the protease domain with its cofactor. The most potent compound demonstrated efficient inhibition of ZIKV propagation in vitro in human fetal neural progenitor cells and in vivo in SJL mice. The inhibitory scaffolds could be further developed into valuable research reagents and, ultimately, provide a roadmap for the selection of efficient inhibitors of ZIKV infection.
Collapse
|
268
|
Sánchez-Carrera D, Bravo-Navas S, Cabezón E, Arechaga I, Cabezas M, Yáñez L, Pipaón C. Fludarabine resistance mediated by aminoglycoside-3'-phosphotransferase-IIa and the structurally related eukaryotic cAMP-dependent protein kinase. FASEB J 2017; 31:3007-3017. [PMID: 28373209 DOI: 10.1096/fj.201601245r] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 03/13/2017] [Indexed: 11/11/2022]
Abstract
While working with G418-resistant stably transfected cells, we realized the neomycin resistance (NeoR) gene, which encodes the aminoglycoside-3'-phosphotransferase-IIa [APH(3')-IIa], also confers resistance to the nucleoside analog fludarabine. Fludarabine is a cytostatic drug widely used in the treatment of hematologic and solid tumors, as well as in the conditioning of patients before transplantation of hematopoietic progenitors. We present evidence that NeoR-transfected cells do not incorporate fludarabine, thus avoiding DNA damage caused by the drug, evidenced by a lack of FANCD2 monoubiquitination and impaired apoptosis. A screening of other nucleoside analogs revealed that APH(3')-IIa only protects against ATP purine analogs. Moreover, APH(3')-IIa ATPase activity is inhibited by fludarabine monophosphate, suggesting that APH(3')-IIa blocks fludarabine incorporation into DNA by dephosphorylating its active fludarabine triphosphate form. Furthermore, overexpression of the catalytic subunit of the eukaryotic kinase PKA, which is structurally related to APHs, also provides resistance to fludarabine, anticipating its putative utility as a response marker to the drug. Our results preclude the use of Neo marker plasmids in the study of purine analogs and unveils a new resistance mechanism against these chemotherapeuticals.-Sánchez-Carrera, D., Bravo-Navas, S., Cabezón, E., Arechaga, I., Cabezas, M., Yáñez, L., Pipaón, C. Fludarabine resistance mediated by aminoglycoside-3'-phosphotransferase-IIa and the structurally related eukaryotic cAMP-dependent protein kinase.
Collapse
Affiliation(s)
- Dámaso Sánchez-Carrera
- Laboratorio de Hematología Molecular, Servicio de Hematología y Hemoterapia, Hospital Universitario Marqués de Valdecilla-Instituto de Investigación Marqués de Valdecilla (IDIVAL), Santander, Spain
| | - Sara Bravo-Navas
- Laboratorio de Hematología Molecular, Servicio de Hematología y Hemoterapia, Hospital Universitario Marqués de Valdecilla-Instituto de Investigación Marqués de Valdecilla (IDIVAL), Santander, Spain
| | - Elena Cabezón
- Departamento de Biología Molecular and Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC), Universidad de Cantabria-Consejo Superior de Investigaciones Cientificas (CSIC), Santander, Spain
| | - Ignacio Arechaga
- Departamento de Biología Molecular and Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC), Universidad de Cantabria-Consejo Superior de Investigaciones Cientificas (CSIC), Santander, Spain
| | - Matilde Cabezas
- Departamento de Biología Molecular and Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC), Universidad de Cantabria-Consejo Superior de Investigaciones Cientificas (CSIC), Santander, Spain
| | - Lucrecia Yáñez
- Laboratorio de Hematología Molecular, Servicio de Hematología y Hemoterapia, Hospital Universitario Marqués de Valdecilla-Instituto de Investigación Marqués de Valdecilla (IDIVAL), Santander, Spain
| | - Carlos Pipaón
- Laboratorio de Hematología Molecular, Servicio de Hematología y Hemoterapia, Hospital Universitario Marqués de Valdecilla-Instituto de Investigación Marqués de Valdecilla (IDIVAL), Santander, Spain;
| |
Collapse
|
269
|
Mahdi LK, Higgins MA, Day CJ, Tiralongo J, Hartley-Tassell LE, Jennings MP, Gordon DL, Paton AW, Paton JC, Ogunniyi AD. The Pneumococcal Alpha-Glycerophosphate Oxidase Enhances Nasopharyngeal Colonization through Binding to Host Glycoconjugates. EBioMedicine 2017; 18:236-243. [PMID: 28330602 PMCID: PMC5405170 DOI: 10.1016/j.ebiom.2017.03.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 02/20/2017] [Accepted: 03/02/2017] [Indexed: 11/22/2022] Open
Abstract
Streptococcus pneumoniae (the pneumococcus) is a major human pathogen, causing a broad spectrum of diseases including otitis media, pneumonia, bacteraemia and meningitis. Here we examined the role of a potential pneumococcal meningitis vaccine antigen, alpha-glycerophosphate oxidase (SpGlpO), in nasopharyngeal colonization. We found that serotype 4 and serotype 6A strains deficient in SpGlpO have significantly reduced capacity to colonize the nasopharynx of mice, and were significantly defective in adherence to human nasopharyngeal carcinoma cells in vitro. We also demonstrate that intranasal immunization with recombinant SpGlpO significantly protects mice against subsequent nasal colonization by wild type serotype 4 and serotype 6A strains. Furthermore, we show that SpGlpO binds strongly to lacto/neolacto/ganglio host glycan structures containing the GlcNAcβ1-3Galβ disaccharide, suggesting that SpGlpO enhances colonization of the nasopharynx through its binding to host glycoconjugates. We propose that SpGlpO is a promising vaccine candidate against pneumococcal carriage, and warrants inclusion in a multi-component protein vaccine formulation that can provide robust, serotype-independent protection against all forms of pneumococcal disease.
Collapse
Affiliation(s)
- Layla K Mahdi
- Research Centre for Infectious Diseases, Department of Molecular and Cellular Biology, School of Biological Sciences, The University of Adelaide, SA 5005, Australia
| | - Melanie A Higgins
- Research Centre for Infectious Diseases, Department of Molecular and Cellular Biology, School of Biological Sciences, The University of Adelaide, SA 5005, Australia
| | - Christopher J Day
- Institute For Glycomics, Griffith University, Gold Coast, QLD, 4222, Australia
| | - Joe Tiralongo
- Institute For Glycomics, Griffith University, Gold Coast, QLD, 4222, Australia
| | | | - Michael P Jennings
- Institute For Glycomics, Griffith University, Gold Coast, QLD, 4222, Australia
| | - David L Gordon
- Department of Microbiology and Infectious Diseases, Flinders University, Bedford Park, SA 5042, Australia
| | - Adrienne W Paton
- Research Centre for Infectious Diseases, Department of Molecular and Cellular Biology, School of Biological Sciences, The University of Adelaide, SA 5005, Australia
| | - James C Paton
- Research Centre for Infectious Diseases, Department of Molecular and Cellular Biology, School of Biological Sciences, The University of Adelaide, SA 5005, Australia.
| | - Abiodun D Ogunniyi
- Research Centre for Infectious Diseases, Department of Molecular and Cellular Biology, School of Biological Sciences, The University of Adelaide, SA 5005, Australia.
| |
Collapse
|
270
|
Munir I, Ajmal S, Shah MR, Ahmad A, Hameed A, Ali SA. Protein-drug nanoconjugates: Finding the alternative proteins as drug carrier. Int J Biol Macromol 2017; 101:131-145. [PMID: 28327425 DOI: 10.1016/j.ijbiomac.2017.03.095] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 02/27/2017] [Accepted: 03/13/2017] [Indexed: 02/07/2023]
Abstract
Present study was conducted to establish the interaction of bovine fetuin-A to validate its binding modalities with doxorubicin (Dox). Fetuin-A was purified to highest purity and monodispersity. Green synthesis of fetuin-A conjugated gold nanoparticles (F-GNPs) has been performed giving typical UV-maxima with subtle variation in fourier transform infrared spectroscopy (FTIR). Atomic force microscopy (AFM) revealed spherical shaped, polydisperse F-GNPs of varying sizes, complementing the radius of hydration (19.5-62.4nm) by dynamic light scattering (DLS). Circular dichroism (CD) analysis of fetuin-A with respect to Dox interaction shows remarkable reduction in ellipticity with increasing concentrations of Dox (20-120μM). Fetuin-A:Dox and F-GNPs:Dox at variable concentrations revealed significantly enhanced absorption spectra, while a continuous decrease in florescence (560nm). This effect was more drastic when Dox interact with fetuin-A as compared to F-GNPs. Some known antimicrobial drugs were also investigated under similar conditions, giving strong quenching effect in a dose dependent manner suggesting the significant yet differential interactions. In cytotoxicity assay, fetuin-A:Dox conjugates revealed less toxicity as compared to F-GNPs:Dox and Dox alone. In-silico studies of the fetuin-A:Dox complex suggest that the drug binds in the major grove between beta-sheet and long loop region of D1 domain and stabilized by several hydrogen bonds.
Collapse
Affiliation(s)
- Iqra Munir
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences (ICCBS), University of Karachi, Karachi, 75270, Pakistan.
| | - Sadia Ajmal
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences (ICCBS), University of Karachi, Karachi, 75270, Pakistan.
| | - Muhammad Raza Shah
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences (ICCBS), University of Karachi, Karachi, 75270, Pakistan.
| | - Aftab Ahmad
- School of Pharmacy, Chapman University, 9401 Jeronimo Road, Irvine, CA, 92618, USA.
| | - Abdul Hameed
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences (ICCBS), University of Karachi, Karachi, 75270, Pakistan.
| | - Syed Abid Ali
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences (ICCBS), University of Karachi, Karachi, 75270, Pakistan.
| |
Collapse
|
271
|
Smith CE, Soti S, Jones TA, Nakagawa A, Xue D, Yin H. Non-steroidal Anti-inflammatory Drugs Are Caspase Inhibitors. Cell Chem Biol 2017; 24:281-292. [PMID: 28238723 PMCID: PMC5357154 DOI: 10.1016/j.chembiol.2017.02.003] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 01/07/2017] [Accepted: 01/31/2017] [Indexed: 01/07/2023]
Abstract
Non-steroidal anti-inflammatory drugs (NSAIDs) are among the most commonly used drugs in the world. While the role of NSAIDs as cyclooxygenase (COX) inhibitors is well established, other targets may contribute to anti-inflammation. Here we report caspases as a new pharmacological target for NSAID family drugs such as ibuprofen, naproxen, and ketorolac at physiologic concentrations both in vitro and in vivo. We characterize caspase activity in both in vitro and in cell culture, and combine computational modeling and biophysical analysis to determine the mechanism of action. We observe that inhibition of caspase catalysis reduces cell death and the generation of pro-inflammatory cytokines. Further, NSAID inhibition of caspases is COX independent, representing a new anti-inflammatory mechanism. This finding expands upon existing NSAID anti-inflammatory behaviors, with implications for patient safety and next-generation drug design.
Collapse
Affiliation(s)
- Christina E Smith
- Department of Chemistry & Biochemistry and the BioFrontiers Institute, University of Colorado, Boulder, CO 80309, USA
| | - Subada Soti
- Department of Chemistry & Biochemistry and the BioFrontiers Institute, University of Colorado, Boulder, CO 80309, USA
| | - Torey A Jones
- Department of Chemistry & Biochemistry and the BioFrontiers Institute, University of Colorado, Boulder, CO 80309, USA
| | - Akihisa Nakagawa
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, CO 80309, USA
| | - Ding Xue
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, CO 80309, USA
| | - Hang Yin
- Department of Chemistry & Biochemistry and the BioFrontiers Institute, University of Colorado, Boulder, CO 80309, USA.
| |
Collapse
|
272
|
Kalyvianaki K, Gebhart V, Peroulis N, Panagiotopoulou C, Kiagiadaki F, Pediaditakis I, Aivaliotis M, Moustou E, Tzardi M, Notas G, Castanas E, Kampa M. Antagonizing effects of membrane-acting androgens on the eicosanoid receptor OXER1 in prostate cancer. Sci Rep 2017; 7:44418. [PMID: 28290516 PMCID: PMC5349529 DOI: 10.1038/srep44418] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Accepted: 02/07/2017] [Indexed: 02/08/2023] Open
Abstract
Accumulating evidence during the last decades revealed that androgen can exert membrane initiated actions that involve signaling via specific kinases and the modulation of significant cellular processes, important for prostate cancer cell growth and metastasis. Results of the present work clearly show that androgens can specifically act at the membrane level via the GPCR oxoeicosanoid receptor 1 (OXER1) in prostate cancer cells. In fact, OXER1 expression parallels that of membrane androgen binding in prostate cancer cell lines and tumor specimens, while in silico docking simulation of OXER1 showed that testosterone could bind to OXER1 within the same grove as 5-OxoETE, the natural ligand of OXER1. Interestingly, testosterone antagonizes the effects of 5-oxoETE on specific signaling pathways and rapid effects such as actin cytoskeleton reorganization that ultimately can modulate cell migration and metastasis. These findings verify that membrane-acting androgens exert specific effects through an antagonistic interaction with OXER1. Additionally, this interaction between androgen and OXER1, which is an arachidonic acid metabolite receptor expressed in prostate cancer, provides a novel link between steroid and lipid actions and renders OXER1 as new player in the disease. These findings should be taken into account in the design of novel therapeutic approaches in prostate cancer.
Collapse
Affiliation(s)
- Konstantina Kalyvianaki
- Laboratory of Experimental Endocrinology, School of Medicine, University of Crete, Heraklion, GR-71003, Greece
| | | | - Nikolaos Peroulis
- Laboratory of Experimental Endocrinology, School of Medicine, University of Crete, Heraklion, GR-71003, Greece
| | - Christina Panagiotopoulou
- Laboratory of Experimental Endocrinology, School of Medicine, University of Crete, Heraklion, GR-71003, Greece
| | - Fotini Kiagiadaki
- Laboratory of Experimental Endocrinology, School of Medicine, University of Crete, Heraklion, GR-71003, Greece
| | - Iosif Pediaditakis
- Department of Pharmacology, School of Medicine, University of Crete, Heraklion, GR-71003, Greece.,Institute of Molecular Biology &Biotechnology, Foundation of Research &Technology-Hellas (IMBB-FORTH), Heraklion, Greece
| | - Michalis Aivaliotis
- Proteomics Facility at Institute of Molecular Biology and Biotechnology, Foundation of Research and Technology-Hellas, Heraklion, Crete
| | - Eleni Moustou
- Department of Pathology, School of Medicine, University of Crete, Heraklion, GR-71003, Greece
| | - Maria Tzardi
- Department of Pathology, School of Medicine, University of Crete, Heraklion, GR-71003, Greece
| | - George Notas
- Laboratory of Experimental Endocrinology, School of Medicine, University of Crete, Heraklion, GR-71003, Greece
| | - Elias Castanas
- Laboratory of Experimental Endocrinology, School of Medicine, University of Crete, Heraklion, GR-71003, Greece
| | - Marilena Kampa
- Laboratory of Experimental Endocrinology, School of Medicine, University of Crete, Heraklion, GR-71003, Greece
| |
Collapse
|
273
|
Tavassoli Z, Taghdir M, Ranjbar B. Renin inhibition by soyasaponin I: a potent native anti-hypertensive compound. J Biomol Struct Dyn 2017; 36:166-176. [DOI: 10.1080/07391102.2016.1270855] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Zeinab Tavassoli
- Department of Biophysics, Tarbiat Modares University, Tehran, Iran
| | - Majid Taghdir
- Department of Biophysics, Tarbiat Modares University, Tehran, Iran
| | - Bijan Ranjbar
- Department of Biophysics, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
274
|
Dang L, Rougé P, Van Damme EJM. Amaranthin-Like Proteins with Aerolysin Domains in Plants. FRONTIERS IN PLANT SCIENCE 2017; 8:1368. [PMID: 28848572 PMCID: PMC5554364 DOI: 10.3389/fpls.2017.01368] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 07/21/2017] [Indexed: 05/20/2023]
Abstract
Amaranthin is a homodimeric lectin that was first discovered in the seeds of Amaranthus caudatus and serves as a model for the family of amaranthin-like lectins. Though these lectins have been purified and characterized only from plant species belonging to the Amaranthaceae, evidence accumulated in recent years suggests that sequences containing amaranthin domains are widely distributed in plants. In this study, 84 plant genomes have been screened to investigate the distribution of amaranthin domains. A total of 265 sequences with amaranthin domains were retrieved from 34 plant genomes. Within this group of amaranthin homologs, 22 different domain architectures can be distinguished. The most common domain combination consists of two amaranthin domains followed by a domain with sequence similarity to aerolysin. The latter protein belongs to the group of β-pore-forming toxins produced by bacteria such as Aeromonas sp. and exerts its toxicity by making transmembrane pores in the target membrane, as such facilitating bacterial invasion. In addition, amaranthin domains also occur in association with five other protein domains, including the fascin domain, the alpha/beta hydrolase domain, the TRAF-like domain, the B box type zinc finger domain and the Bet v1 domain. All 16 amaranthin-like proteins retrieved from the cucumber genome possess a similar domain architecture consisting of two amaranthin domains linked to one aerolysin domain. Based on phylogenetic differences, four sequences were selected for further investigation. Subcellular localization studies revealed that the amaranthin-like proteins from cucumber reside in the cytoplasm and/or the nucleus. Analyses using qPCR showed that the transcript levels for the amaranthin-like sequences are typically low and expression levels vary among tissues during the development of cucumber plants. Furthermore, the expression of amaranthin-like genes is enhanced after different abiotic stresses, suggesting that these amaranthin-like proteins play a role in the stress response. Finally, molecular modeling was performed to unravel the structure of amaranthin-like proteins and their carbohydrate-binding sites. This study provided valuable information on the distribution, phylogenetic relationships, and possible biological roles of amaranthin-like proteins in plants.
Collapse
Affiliation(s)
- Liuyi Dang
- Laboratory of Biochemistry and Glycobiology, Department of Molecular Biotechnology, Ghent UniversityGhent, Belgium
| | - Pierre Rougé
- UMR 152 PHARMA-DEV, Université de ToulouseToulouse, France
| | - Els J. M. Van Damme
- Laboratory of Biochemistry and Glycobiology, Department of Molecular Biotechnology, Ghent UniversityGhent, Belgium
- *Correspondence: Els J. M. Van Damme,
| |
Collapse
|
275
|
Das A, Bhattacharya S. Different Types of Molecular Docking Based on Variations of Interacting Molecules. PHARMACEUTICAL SCIENCES 2017. [DOI: 10.4018/978-1-5225-1762-7.ch031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Molecular docking plays an important role in drug discovery research by facilitating target identification, target validation, virtual screening for lead identification and lead optimization. Depending upon the nature of the disease of interest, targets can be either protein or DNA while drugs are mostly organic small molecules. Different types of molecular docking techniques like protein-protein or protein-DNA or protein-small molecule or DNA-small molecule are employed for achieving the above mentioned objectives. This chapter provides a clear idea of the position of molecular docking in drug discovery with detailed discussion on different types of molecular docking based on the varieties of interacting partners. Subsequently the authors provide a detailed list of tools that can be used for docking in drug discovery and discus some examples of molecular docking in drug discovery before concluding with a remark on future areas of improvement in molecular docking related to drug discovery.
Collapse
|
276
|
Ribaudo G, Vendrame T, Bova S. Isoflavones from Maclura pomifera: structural elucidation and in silico evaluation of their interaction with PDE5. Nat Prod Res 2016; 31:1988-1994. [DOI: 10.1080/14786419.2016.1269101] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Giovanni Ribaudo
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | | | - Sergio Bova
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| |
Collapse
|
277
|
Abstract
We developed a hybrid quantum mechanical/molecular mechanical (QM/MM) on-the-fly docking algorithm to address the challenges of treating polarization and selected metal interactions in docking. The algorithm is based on our classical docking algorithm Attracting Cavities and relies on the semiempirical self-consistent charge density functional tight-binding (SCC-DFTB) method and the CHARMM force field. We benchmarked the performance of this approach on three very diverse data sets: (1) the Astex Diverse set of 85 common noncovalent drug/target complexes formed both by hydrophobic and electrostatic interactions; (2) a zinc metalloprotein data set of 281 complexes, where polarization is strong and ligand/protein interactions are dominated by electrostatic interactions; and (3) a heme protein data set of 72 complexes, where ligand/protein interactions are dominated by covalent ligand/iron binding. Redocking performance of the on-the-fly QM/MM docking algorithm was compared to the performance of classical Attracting Cavities, AutoDock, AutoDock Vina, and GOLD. The results demonstrate that the QM/MM code preserves the high accuracy of most classical scores on the Astex Diverse set, while it yields significant improvements on both sets of metalloproteins at moderate computational cost.
Collapse
Affiliation(s)
- Prasad Chaskar
- SIB Swiss Institute of Bioinformatics , Molecular Modeling Group, CH-1015 Lausanne, Switzerland
| | - Vincent Zoete
- SIB Swiss Institute of Bioinformatics , Molecular Modeling Group, CH-1015 Lausanne, Switzerland
| | - Ute F Röhrig
- SIB Swiss Institute of Bioinformatics , Molecular Modeling Group, CH-1015 Lausanne, Switzerland
| |
Collapse
|
278
|
Computational Discovery of Putative Leads for Drug Repositioning through Drug-Target Interaction Prediction. PLoS Comput Biol 2016; 12:e1005219. [PMID: 27893735 PMCID: PMC5125559 DOI: 10.1371/journal.pcbi.1005219] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 10/21/2016] [Indexed: 12/23/2022] Open
Abstract
De novo experimental drug discovery is an expensive and time-consuming task. It requires the identification of drug-target interactions (DTIs) towards targets of biological interest, either to inhibit or enhance a specific molecular function. Dedicated computational models for protein simulation and DTI prediction are crucial for speed and to reduce the costs associated with DTI identification. In this paper we present a computational pipeline that enables the discovery of putative leads for drug repositioning that can be applied to any microbial proteome, as long as the interactome of interest is at least partially known. Network metrics calculated for the interactome of the bacterial organism of interest were used to identify putative drug-targets. Then, a random forest classification model for DTI prediction was constructed using known DTI data from publicly available databases, resulting in an area under the ROC curve of 0.91 for classification of out-of-sampling data. A drug-target network was created by combining 3,081 unique ligands and the expected ten best drug targets. This network was used to predict new DTIs and to calculate the probability of the positive class, allowing the scoring of the predicted instances. Molecular docking experiments were performed on the best scoring DTI pairs and the results were compared with those of the same ligands with their original targets. The results obtained suggest that the proposed pipeline can be used in the identification of new leads for drug repositioning. The proposed classification model is available at http://bioinformatics.ua.pt/software/dtipred/. The emergence of multi-resistant bacterial strains and the existing void in the discovery and development of new classes of antibiotics is a growing concern. Indeed, some bacterial strains are now resistant to last-line antibiotics and considered untreatable. Drug repositioning has been suggested as a strategy to minimize time and cost expenses until the drug reaches the market, compared to traditional drug design. Drug-target interactions (DTIs) are the basis of rational drug design and thus, we proposed a computational approach to predict DTIs solely based on the primary sequence of the protein and the simplified molecular-input line-entry system of the ligand. In addition, network metrics are used to identify vital putative drug-targets in bacteria. Molecular docking experiments were performed to compare the binding affinities between a given ligand and a putative drug-target, as well as with their original targets. According to the docking results, the predicted DTIs have better or similar binding activities than the ligand and their real target, indicating the validity of the proposed model.
Collapse
|
279
|
Berndsen CE, Young BH, McCormick QJ, Enke RA. Connecting common genetic polymorphisms to protein function: A modular project sequence for lecture or lab. BIOCHEMISTRY AND MOLECULAR BIOLOGY EDUCATION : A BIMONTHLY PUBLICATION OF THE INTERNATIONAL UNION OF BIOCHEMISTRY AND MOLECULAR BIOLOGY 2016; 44:526-536. [PMID: 27277577 DOI: 10.1002/bmb.20976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2015] [Revised: 03/12/2016] [Accepted: 04/03/2016] [Indexed: 06/06/2023]
Abstract
Single nucleotide polymorphisms (SNPs) in DNA can result in phenotypes where the biochemical basis may not be clear due to the lack of protein structures. With the growing number of modeling and simulation software available on the internet, students can now participate in determining how small changes in genetic information impact cellular protein structure and function. We have developed a modular series of activities to engage lab or lecture students in examining the basis for common phenotypes. The activities range from basic phenotype testing/observation to DNA sequencing and simulation of protein structure and dynamics. We provide as an example study of the bitterness receptor TAS2R38 and PTC tasting, however these activities are applicable to other SNPs or genomic variants with a direct connection to an observable phenotype. These activities are modular and can be mixed to meet the student capabilities and infrastructure availability. The complete sequence of activities will demonstrate the direct connection between gene structure, protein structure and organism function. © 2016 by The International Union of Biochemistry and Molecular Biology, 44(6):526-536, 2016.
Collapse
Affiliation(s)
- Christopher E Berndsen
- Department of Chemistry and Biochemistry, James Madison University, Virginia
- Center for Genome and Metagenome Studies, James Madison University, Virginia
| | - Byron H Young
- Department of Chemistry and Biochemistry, James Madison University, Virginia
| | | | - Raymond A Enke
- Center for Genome and Metagenome Studies, James Madison University, Virginia
- Department of Biology, James Madison University, Virginia
| |
Collapse
|
280
|
Solomonov AV, Shipitsyna MK, Vashurin AS, Rumyantsev EV, Timin AS, Ivanov SP. Analysis of binding ability of two tetramethylpyridylporphyrins to albumin and its complex with bilirubin. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2016; 168:12-20. [PMID: 27267279 DOI: 10.1016/j.saa.2016.05.044] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 05/27/2016] [Accepted: 05/28/2016] [Indexed: 06/06/2023]
Abstract
An interaction between 5,10,15,20-tetrakis-(N-methyl-x-pyridyl)porphyrins, x=2; 4 (TMPyPs) with bovine serum albumin (BSA) and its bilirubin (BR) complex was investigated by UV-Viz and fluorescence spectroscopy under imitated physiological conditions involving molecular docking studies. The parameters of forming intermolecular complexes (binding constants, quenching rate constants, quenching sphere radius etc.) were determined. It was showed that the interaction between proteins and TMPyPs occurs via static quenching of protein fluorescence and has predominantly hydrophobic and electrostatic character. It was revealed that obtained complexes are relatively stable, but in the case of TMPyP4 binding with proteins occurs better than TMPyP2. Nevertheless, both TMPyPs have better binding ability with free protein compared to BRBSA at the same time. The influence of TMPyPs on the conformational changes in protein molecules was studied using synchronous fluorescence spectroscopy. It was found that there is no competition of BR with TMPyPs for binging sites on protein molecule and BR displacement does not occur. Molecular docking calculations have showed that TMPyPs can bind with albumin via tryptophan residue in the hydrophilic binding site of protein molecule but it is not one possible interaction way.
Collapse
Affiliation(s)
- Alexey V Solomonov
- Inorganic Chemistry Department, Ivanovo State University of Chemistry and Technology, 7 Sheremetevskij prosp., 153000 Ivanovo, Russian Federation; Department of Materials and Interfaces, Weizmann Institute of Science, Rehovot 7610001, Israel.
| | - Maria K Shipitsyna
- Inorganic Chemistry Department, Ivanovo State University of Chemistry and Technology, 7 Sheremetevskij prosp., 153000 Ivanovo, Russian Federation.
| | - Arthur S Vashurin
- Inorganic Chemistry Department, Ivanovo State University of Chemistry and Technology, 7 Sheremetevskij prosp., 153000 Ivanovo, Russian Federation.
| | - Evgeniy V Rumyantsev
- Inorganic Chemistry Department, Ivanovo State University of Chemistry and Technology, 7 Sheremetevskij prosp., 153000 Ivanovo, Russian Federation.
| | - Alexander S Timin
- Inorganic Chemistry Department, Ivanovo State University of Chemistry and Technology, 7 Sheremetevskij prosp., 153000 Ivanovo, Russian Federation; Tomsk Polytechnic University, RASA Center in Tomsk, 30, Lenin Avenue, 634500 Tomsk, Russian Federation.
| | - Sergey P Ivanov
- Ufa Institute of Chemistry of the Russian Academy of Sciences, Prosp. Oktyabrya 71, Ufa 450045, Russian Federation.
| |
Collapse
|
281
|
In Silico Approaches to Identify Mutagenesis Targets to Probe and Alter Protein-Cofactor and Protein-Protein Functional Relationships. Methods Mol Biol 2016; 1498:181-190. [PMID: 27709576 DOI: 10.1007/978-1-4939-6472-7_12] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2023]
Abstract
When performing site-directed mutagenesis experiments to study protein structure-function relationships, ideally one would know the structure of the protein under study. It is also very useful to have structures of multiple related proteins in order to determine whether or not particular amino acid residues are conserved in the structures either in the active site of an enzyme at the surface of a protein or at a putative protein-protein interface. While many protein structures are available in the Protein Data Base (PDB), a structure of the protein of interest may not be available. In the study of reversible and often transient protein-protein interactions it is rare to have a structure of the complex of the two interacting proteins. In this chapter, methods are described for comparing protein structures, generating putative structures of proteins with homology models based on the protein primary sequence, and generating docking models to predict interaction sites between proteins and cofactor-protein interactions. The rationale used to predict mutagenesis targets from these structures and models is also described.
Collapse
|
282
|
Wagner AE, Piegholdt S, Rabe D, Baenas N, Schloesser A, Eggersdorfer M, Stocker A, Rimbach G. Epigallocatechin gallate affects glucose metabolism and increases fitness and lifespan in Drosophila melanogaster. Oncotarget 2016; 6:30568-78. [PMID: 26375250 PMCID: PMC4741552 DOI: 10.18632/oncotarget.5215] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 08/28/2015] [Indexed: 11/25/2022] Open
Abstract
In this study, we tested whether a standardized epigallocatechin-3-gallate (EGCG) rich green tea extract (comprising > 90% EGCG) affects fitness and lifespan as well as parameters of glucose metabolism and energy homeostasis in the fruit fly, Drosophila melanogaster. Following the application of the green tea extract a significant increase in the mean lifespan (+ 3.3 days) and the 50% survival (+ 4.3 days) as well as improved fitness was detected. These effects went along an increased expression of Spargel, the homolog of mammalian PGC1α, which has been reported to affect lifespan in flies. Intriguingly, in flies, treatment with the green tea extract decreased glucose concentrations, which were accompanied by an inhibition of α-amylase and α-glucosidase activity. Computational docking analysis proved the potential of EGCG to dock into the substrate binding pocket of α-amylase and to a greater extent into α-glucosidase. Furthermore, we demonstrate that EGCG downregulates insulin-like peptide 5 and phosphoenolpyruvate carboxykinase, major regulators of glucose metabolism, as well as the Drosophila homolog of leptin, unpaired 2. We propose that a decrease in glucose metabolism in connection with an upregulated expression of Spargel contribute to the better fitness and the extended lifespan in EGCG-treated flies.
Collapse
Affiliation(s)
- Anika E Wagner
- Institute of Human Nutrition and Food Science, University of Kiel, Kiel, Germany
| | - Stefanie Piegholdt
- Institute of Human Nutrition and Food Science, University of Kiel, Kiel, Germany
| | - Doerte Rabe
- Institute of Human Nutrition and Food Science, University of Kiel, Kiel, Germany
| | - Nieves Baenas
- Institute of Human Nutrition and Food Science, University of Kiel, Kiel, Germany.,Centro de Edafología y Biología Aplicada del Segura (CEBAS-CSIC), Campus Universitario de Espinardo, Murcia, Spain
| | - Anke Schloesser
- Institute of Human Nutrition and Food Science, University of Kiel, Kiel, Germany
| | | | - Achim Stocker
- Department of Chemistry and Biochemistry, University of Bern, Bern, Switzerland
| | - Gerald Rimbach
- Institute of Human Nutrition and Food Science, University of Kiel, Kiel, Germany
| |
Collapse
|
283
|
de Barros NB, Macedo SR, Ferreira AS, Tagliari MP, Zanchi FB, Kayano AM, Soares AM, Nicolete R. Liposomes containing an ASP49-phospholipase A 2 from Bothrops jararacussu snake venom as experimental therapy against cutaneous leishmaniasis. Int Immunopharmacol 2016; 36:225-231. [DOI: 10.1016/j.intimp.2016.04.025] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Revised: 04/05/2016] [Accepted: 04/18/2016] [Indexed: 10/21/2022]
|
284
|
Hu D, Sheng Z, Gao G, Siu F, Liu C, Wan Q, Gong P, Zheng H, Ma Y, Cai L. Activatable albumin-photosensitizer nanoassemblies for triple-modal imaging and thermal-modulated photodynamic therapy of cancer. Biomaterials 2016; 93:10-19. [DOI: 10.1016/j.biomaterials.2016.03.037] [Citation(s) in RCA: 118] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Revised: 03/21/2016] [Accepted: 03/28/2016] [Indexed: 01/12/2023]
|
285
|
Simigdala N, Gao Q, Pancholi S, Roberg-Larsen H, Zvelebil M, Ribas R, Folkerd E, Thompson A, Bhamra A, Dowsett M, Martin LA. Cholesterol biosynthesis pathway as a novel mechanism of resistance to estrogen deprivation in estrogen receptor-positive breast cancer. Breast Cancer Res 2016; 18:58. [PMID: 27246191 PMCID: PMC4888666 DOI: 10.1186/s13058-016-0713-5] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 04/26/2016] [Indexed: 02/07/2023] Open
Abstract
Background Therapies targeting estrogenic stimulation in estrogen receptor-positive (ER+) breast cancer (BC) reduce mortality, but resistance remains a major clinical problem. Molecular studies have shown few high-frequency mutations to be associated with endocrine resistance. In contrast, expression profiling of primary ER+ BC samples has identified several promising signatures/networks for targeting. Methods To identify common adaptive mechanisms associated with resistance to aromatase inhibitors (AIs), we assessed changes in global gene expression during adaptation to long-term estrogen deprivation (LTED) in a panel of ER+ BC cell lines cultured in 2D on plastic (MCF7, T47D, HCC1428, SUM44 and ZR75.1) or in 3D on collagen (MCF7) to model the stromal compartment. Furthermore, dimethyl labelling followed by LC-MS/MS was used to assess global changes in protein abundance. The role of target genes/proteins on proliferation, ER-mediated transcription and recruitment of ER to target gene promoters was analysed. Results The cholesterol biosynthesis pathway was the common upregulated pathway in the ER+ LTED but not the ER– LTED cell lines, suggesting a potential mechanism dependent on continued ER expression. Targeting the individual genes of the cholesterol biosynthesis pathway with siRNAs caused a 30–50 % drop in proliferation. Further analysis showed increased expression of 25-hydroxycholesterol (HC) in the MCF7 LTED cells. Exogenous 25-HC or 27-HC increased ER-mediated transcription and expression of the endogenous estrogen-regulated gene TFF1 in ER+ LTED cells but not in the ER– LTED cells. Additionally, recruitment of the ER and CREB-binding protein (CBP) to the TFF1 and GREB1 promoters was increased upon treatment with 25-HC and 27-HC. In-silico analysis of two independent studies of primary ER+ BC patients treated with neoadjuvant AIs showed that increased expression of MSMO1, EBP, LBR and SQLE enzymes, required for cholesterol synthesis and increased in our in-vitro models, was significantly associated with poor response to endocrine therapy. Conclusion Taken together, these data provide support for the role of cholesterol biosynthesis enzymes and the cholesterol metabolites, 25-HC and 27-HC, in a novel mechanism of resistance to endocrine therapy in ER+ BC that has potential as a therapeutic target. Electronic supplementary material The online version of this article (doi:10.1186/s13058-016-0713-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Nikiana Simigdala
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Qiong Gao
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Sunil Pancholi
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | | | - Marketa Zvelebil
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Ricardo Ribas
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Elizabeth Folkerd
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK.,Academic Department of Biochemistry, Royal Marsden Hospital, London, SW3 6JJ, UK
| | - Andrew Thompson
- Proteomics Core Facility, Institute of Cancer Research, London, SW3 6JB, UK
| | - Amandeep Bhamra
- Proteomics Core Facility, Institute of Cancer Research, London, SW3 6JB, UK
| | - Mitch Dowsett
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK.,Academic Department of Biochemistry, Royal Marsden Hospital, London, SW3 6JJ, UK
| | - Lesley-Ann Martin
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK.
| |
Collapse
|
286
|
Mumcuoglu M, Gurkan-Alp AS, Buyukbingol E, Cetin-Atalay R. Retinoid N-(1H-benzo[d]imidazol-2-yl)-5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalene-2-carboxamide induces p21-dependent senescence in breast cancer cells. Steroids 2016; 108:31-8. [PMID: 26898539 DOI: 10.1016/j.steroids.2016.02.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2015] [Revised: 01/20/2016] [Accepted: 02/11/2016] [Indexed: 12/27/2022]
Abstract
Retinoids have been implicated as pharmacological agents for the prevention and treatment of various types of cancers, including breast cancers. We analyzed 27 newly synthesized retinoids for their bioactivity on breast, liver, and colon cancer cells. Majority of the retinoids demonstrated selective bioactivity on breast cancer cells. Retinoid 17 had a significant inhibitory activity (IC50 3.5 μM) only on breast cancer cells while no growth inhibition observed with liver and colon cancer cells. The breast cancer selective growth inhibitory action by retinoid 17 was defined as p21-dependent cell death, reminiscent of senescence, which is an indicator of targeted receptor mediated bioactivity. A comparative analysis of retinoid receptor gene expression levels in different breast cancer cells and IC50 values of 17 indicated the involvement of Retinoid X receptors in the cytotoxic bioactivity of retinoid 17 in the senescence associated cell death. Furthermore, siRNA knockdown studies with RXRγ induced decrease in cell proliferation. Therefore, we suggest that retinoid derivatives that target RXRγ, can be considered for breast cancer therapies.
Collapse
Affiliation(s)
- Mine Mumcuoglu
- LOSEV the Foundation for Children with Leukemia, Cancer Genetics Research Laboratory, Ankara, Turkey; Department of Molecular Biology and Genetics, Bilkent University, Bilkent, 06800 Ankara, Turkey
| | - A Selen Gurkan-Alp
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Ankara University, Turkey
| | - Erdem Buyukbingol
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Ankara University, Turkey
| | - Rengul Cetin-Atalay
- LOSEV the Foundation for Children with Leukemia, Cancer Genetics Research Laboratory, Ankara, Turkey; Graduate School of Informatics, Cancer Systems Biology Laboratory, METU, 06800 Ankara, Turkey.
| |
Collapse
|
287
|
Mallon M, Dutt S, Schrader T, Crowley PB. Protein Camouflage: Supramolecular Anion Recognition by Ubiquitin. Chembiochem 2016; 17:774-83. [PMID: 26818656 DOI: 10.1002/cbic.201500477] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Indexed: 11/11/2022]
Abstract
Progress in the field of bio-supramolecular chemistry, the bottom-up assembly of protein-ligand systems, relies on a detailed knowledge of molecular recognition. To address this issue, we have characterised complex formation between human ubiquitin (HUb) and four supramolecular anions. The ligands were: pyrenetetrasulfonic acid (4PSA), p-sulfonato-calix[4]arene (SCLX4), bisphosphate tweezers (CLR01) and meso-tetrakis (4-sulfonatophenyl)porphyrin (TPPS), which vary in net charge, size, shape and hydrophobicity. All four ligands induced significant changes in the HSQC spectrum of HUb. Chemical shift perturbations and line-broadening effects were used to identify binding sites and to quantify affinities. Supporting data were obtained from docking simulations. It was found that these weakly interacting ligands bind to extensive surface patches on HUb. A comparison of the data suggests some general indicators for the protein-binding specificity of supramolecular anions. Differences in binding were observed between the cavity-containing and planar ligands. The former had a preference for the arginine-rich, flexible C terminus of HUb.
Collapse
Affiliation(s)
- Madeleine Mallon
- School of Chemistry, National University of Ireland Galway, University Road, Galway, Ireland
| | - Som Dutt
- Institute for Organic Chemistry, University of Duisburg-Essen, Universitätsstrasse 7, 45117, Essen, Germany
| | - Thomas Schrader
- Institute for Organic Chemistry, University of Duisburg-Essen, Universitätsstrasse 7, 45117, Essen, Germany
| | - Peter B Crowley
- School of Chemistry, National University of Ireland Galway, University Road, Galway, Ireland.
| |
Collapse
|
288
|
Application of PEG-400 as a green biodegradable polymeric medium for the catalyst-free synthesis of spiro-dihydropyridines and their use as acetyl and butyrylcholinesterase inhibitors. Bioorg Med Chem 2016; 24:1408-17. [DOI: 10.1016/j.bmc.2016.02.019] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2016] [Revised: 02/07/2016] [Accepted: 02/09/2016] [Indexed: 11/23/2022]
|
289
|
Glaab E. Building a virtual ligand screening pipeline using free software: a survey. Brief Bioinform 2016; 17:352-66. [PMID: 26094053 PMCID: PMC4793892 DOI: 10.1093/bib/bbv037] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Revised: 05/20/2015] [Indexed: 12/17/2022] Open
Abstract
Virtual screening, the search for bioactive compounds via computational methods, provides a wide range of opportunities to speed up drug development and reduce the associated risks and costs. While virtual screening is already a standard practice in pharmaceutical companies, its applications in preclinical academic research still remain under-exploited, in spite of an increasing availability of dedicated free databases and software tools. In this survey, an overview of recent developments in this field is presented, focusing on free software and data repositories for screening as alternatives to their commercial counterparts, and outlining how available resources can be interlinked into a comprehensive virtual screening pipeline using typical academic computing facilities. Finally, to facilitate the set-up of corresponding pipelines, a downloadable software system is provided, using platform virtualization to integrate pre-installed screening tools and scripts for reproducible application across different operating systems.
Collapse
|
290
|
Zoete V, Schuepbach T, Bovigny C, Chaskar P, Daina A, Röhrig UF, Michielin O. Attracting cavities for docking. Replacing the rough energy landscape of the protein by a smooth attracting landscape. J Comput Chem 2016; 37:437-47. [PMID: 26558715 PMCID: PMC4738475 DOI: 10.1002/jcc.24249] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 09/09/2015] [Accepted: 10/11/2015] [Indexed: 01/24/2023]
Abstract
Molecular docking is a computational approach for predicting the most probable position of ligands in the binding sites of macromolecules and constitutes the cornerstone of structure-based computer-aided drug design. Here, we present a new algorithm called Attracting Cavities that allows molecular docking to be performed by simple energy minimizations only. The approach consists in transiently replacing the rough potential energy hypersurface of the protein by a smooth attracting potential driving the ligands into protein cavities. The actual protein energy landscape is reintroduced in a second step to refine the ligand position. The scoring function of Attracting Cavities is based on the CHARMM force field and the FACTS solvation model. The approach was tested on the 85 experimental ligand-protein structures included in the Astex diverse set and achieved a success rate of 80% in reproducing the experimental binding mode starting from a completely randomized ligand conformer. The algorithm thus compares favorably with current state-of-the-art docking programs.
Collapse
Affiliation(s)
- Vincent Zoete
- Bâtiment Génopode, SIB Swiss Institute of Bioinformatics, Quartier Sorge, CH-1015 Lausanne, Switzerland
| | - Thierry Schuepbach
- Bâtiment Génopode, SIB Swiss Institute of Bioinformatics, Quartier Sorge, CH-1015 Lausanne, Switzerland
| | - Christophe Bovigny
- Bâtiment Génopode, SIB Swiss Institute of Bioinformatics, Quartier Sorge, CH-1015 Lausanne, Switzerland
| | - Prasad Chaskar
- Bâtiment Génopode, SIB Swiss Institute of Bioinformatics, Quartier Sorge, CH-1015 Lausanne, Switzerland
| | - Antoine Daina
- Bâtiment Génopode, SIB Swiss Institute of Bioinformatics, Quartier Sorge, CH-1015 Lausanne, Switzerland
| | - Ute F Röhrig
- Bâtiment Génopode, SIB Swiss Institute of Bioinformatics, Quartier Sorge, CH-1015 Lausanne, Switzerland
| | - Olivier Michielin
- Bâtiment Génopode, SIB Swiss Institute of Bioinformatics, Quartier Sorge, CH-1015 Lausanne, Switzerland
- Ludwig Institute for Cancer Research, Centre Hospitalier Universitaire Vaudois, CH-1011 Lausanne, Switzerland
- Department of Oncology, University of Lausanne and Centre Hospitalier Universitaire Vaudois (CHUV), CH-1011 Lausanne, Switzerland
| |
Collapse
|
291
|
Das A, Bhattacharya S. Different Types of Molecular Docking Based on Variations of Interacting Molecules. METHODS AND ALGORITHMS FOR MOLECULAR DOCKING-BASED DRUG DESIGN AND DISCOVERY 2016. [DOI: 10.4018/978-1-5225-0115-2.ch006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Molecular docking plays an important role in drug discovery research by facilitating target identification, target validation, virtual screening for lead identification and lead optimization. Depending upon the nature of the disease of interest, targets can be either protein or DNA while drugs are mostly organic small molecules. Different types of molecular docking techniques like protein-protein or protein-DNA or protein-small molecule or DNA-small molecule are employed for achieving the above mentioned objectives. This chapter provides a clear idea of the position of molecular docking in drug discovery with detailed discussion on different types of molecular docking based on the varieties of interacting partners. Subsequently the authors provide a detailed list of tools that can be used for docking in drug discovery and discus some examples of molecular docking in drug discovery before concluding with a remark on future areas of improvement in molecular docking related to drug discovery.
Collapse
|
292
|
Woods KN, Pfeffer J. Using THz Spectroscopy, Evolutionary Network Analysis Methods, and MD Simulation to Map the Evolution of Allosteric Communication Pathways in c-Type Lysozymes. Mol Biol Evol 2016; 33:40-61. [PMID: 26337549 PMCID: PMC4693973 DOI: 10.1093/molbev/msv178] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
It is now widely accepted that protein function is intimately tied with the navigation of energy landscapes. In this framework, a protein sequence is not described by a distinct structure but rather by an ensemble of conformations. And it is through this ensemble that evolution is able to modify a protein's function by altering its landscape. Hence, the evolution of protein functions involves selective pressures that adjust the sampling of the conformational states. In this work, we focus on elucidating the evolutionary pathway that shaped the function of individual proteins that make-up the mammalian c-type lysozyme subfamily. Using both experimental and computational methods, we map out specific intermolecular interactions that direct the sampling of conformational states and accordingly, also underlie shifts in the landscape that are directly connected with the formation of novel protein functions. By contrasting three representative proteins in the family we identify molecular mechanisms that are associated with the selectivity of enhanced antimicrobial properties and consequently, divergent protein function. Namely, we link the extent of localized fluctuations involving the loop separating helices A and B with shifts in the equilibrium of the ensemble of conformational states that mediate interdomain coupling and concurrently moderate substrate binding affinity. This work reveals unique insights into the molecular level mechanisms that promote the progression of interactions that connect the immune response to infection with the nutritional properties of lactation, while also providing a deeper understanding about how evolving energy landscapes may define present-day protein function.
Collapse
|
293
|
Lac D, Feng C, Bhardwaj G, Le H, Tran J, Xing L, Fung G, Liu R, Cheng H, Lam KS. Covalent Chemical Ligation Strategy for Mono- and Polyclonal Immunoglobulins at Their Nucleotide Binding Sites. Bioconjug Chem 2015; 27:159-69. [PMID: 26630124 DOI: 10.1021/acs.bioconjchem.5b00574] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Nonspecific ligation methods have been traditionally used to chemically modify immunoglobulins. Site-specific ligation of compounds (toxins or ligands) to antibodies has become increasingly important in the fields of therapeutic antibody-drug conjugates and bispecific antibodies. In this present study, we took advantage of the reported nucleotide-binding pocket (NBP) in the Fab arms of immunoglobulins by developing indole-based, 5-fluoro-2,4-dinitrobenzene-derivatized OBOC peptide libraries for the identification of affinity elements that can be used as site-specific derivatization agents against both mono- and polyclonal antibodies. Ligation can occur at any one of the few lysine residues located at the NBP. Immunoconjugates resulting from such affinity elements can be used as therapeutics against cancer or infectious agents.
Collapse
Affiliation(s)
| | | | | | | | | | - Li Xing
- Department of Molecular & Cellular Biology, University of California, Davis , Davis, California 95616, United States
| | | | | | - Holland Cheng
- Department of Molecular & Cellular Biology, University of California, Davis , Davis, California 95616, United States
| | | |
Collapse
|
294
|
Helmich KE, Pereira JH, Gall DL, Heins RA, McAndrew RP, Bingman C, Deng K, Holland KC, Noguera DR, Simmons BA, Sale KL, Ralph J, Donohue TJ, Adams PD, Phillips GN. Structural Basis of Stereospecificity in the Bacterial Enzymatic Cleavage of β-Aryl Ether Bonds in Lignin. J Biol Chem 2015; 291:5234-46. [PMID: 26637355 PMCID: PMC4777856 DOI: 10.1074/jbc.m115.694307] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Indexed: 11/23/2022] Open
Abstract
Lignin is a combinatorial polymer comprising monoaromatic units that are linked via covalent bonds. Although lignin is a potential source of valuable aromatic chemicals, its recalcitrance to chemical or biological digestion presents major obstacles to both the production of second-generation biofuels and the generation of valuable coproducts from lignin's monoaromatic units. Degradation of lignin has been relatively well characterized in fungi, but it is less well understood in bacteria. A catabolic pathway for the enzymatic breakdown of aromatic oligomers linked via β-aryl ether bonds typically found in lignin has been reported in the bacterium Sphingobium sp. SYK-6. Here, we present x-ray crystal structures and biochemical characterization of the glutathione-dependent β-etherases, LigE and LigF, from this pathway. The crystal structures show that both enzymes belong to the canonical two-domain fold and glutathione binding site architecture of the glutathione S-transferase family. Mutagenesis of the conserved active site serine in both LigE and LigF shows that, whereas the enzymatic activity is reduced, this amino acid side chain is not absolutely essential for catalysis. The results include descriptions of cofactor binding sites, substrate binding sites, and catalytic mechanisms. Because β-aryl ether bonds account for 50–70% of all interunit linkages in lignin, understanding the mechanism of enzymatic β-aryl ether cleavage has significant potential for informing ongoing studies on the valorization of lignin.
Collapse
Affiliation(s)
- Kate E Helmich
- From the Department of Biochemistry, University of Wisconsin, Madison, Wisconsin 53706, the United States Department of Energy Great Lakes Bioenergy Research Center, Wisconsin Energy Institute, University of Wisconsin, Madison, Wisconsin 53726
| | - Jose Henrique Pereira
- the Joint BioEnergy Institute, Emeryville, California 94608, the Physical Biosciences Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720
| | - Daniel L Gall
- the United States Department of Energy Great Lakes Bioenergy Research Center, Wisconsin Energy Institute, University of Wisconsin, Madison, Wisconsin 53726, the Departments of Civil and Environmental Engineering and
| | - Richard A Heins
- the Joint BioEnergy Institute, Emeryville, California 94608, the Biological and Engineering Sciences Center, Sandia National Laboratories, Livermore, California 94551
| | - Ryan P McAndrew
- the Joint BioEnergy Institute, Emeryville, California 94608, the Physical Biosciences Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720
| | - Craig Bingman
- From the Department of Biochemistry, University of Wisconsin, Madison, Wisconsin 53706
| | - Kai Deng
- the Joint BioEnergy Institute, Emeryville, California 94608, the Biological and Engineering Sciences Center, Sandia National Laboratories, Livermore, California 94551
| | - Keefe C Holland
- the Joint BioEnergy Institute, Emeryville, California 94608, the Biological and Engineering Sciences Center, Sandia National Laboratories, Livermore, California 94551
| | - Daniel R Noguera
- the United States Department of Energy Great Lakes Bioenergy Research Center, Wisconsin Energy Institute, University of Wisconsin, Madison, Wisconsin 53726, the Departments of Civil and Environmental Engineering and
| | - Blake A Simmons
- the Joint BioEnergy Institute, Emeryville, California 94608, the Biological and Engineering Sciences Center, Sandia National Laboratories, Livermore, California 94551
| | - Kenneth L Sale
- the Joint BioEnergy Institute, Emeryville, California 94608, the Biological and Engineering Sciences Center, Sandia National Laboratories, Livermore, California 94551
| | - John Ralph
- From the Department of Biochemistry, University of Wisconsin, Madison, Wisconsin 53706, the United States Department of Energy Great Lakes Bioenergy Research Center, Wisconsin Energy Institute, University of Wisconsin, Madison, Wisconsin 53726
| | - Timothy J Donohue
- the United States Department of Energy Great Lakes Bioenergy Research Center, Wisconsin Energy Institute, University of Wisconsin, Madison, Wisconsin 53726, Bacteriology, University of Wisconsin, Madison, Wisconsin 53706,
| | - Paul D Adams
- the Joint BioEnergy Institute, Emeryville, California 94608, the Physical Biosciences Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, the Department of Bioengineering, University of California, Berkeley, California 94720, and
| | - George N Phillips
- the Department of Biochemistry and Cell Biology, Rice University, Houston, Texas 77251
| |
Collapse
|
295
|
Sato M, Sakurai N, Suzuki H, Shibata D, Kino K. Enzymatic carboxylation of hydroxystilbenes by the γ-resorcylic acid decarboxylase from Rhizobium radiobacter WU-0108 under reverse reaction conditions. ACTA ACUST UNITED AC 2015. [DOI: 10.1016/j.molcatb.2015.10.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
296
|
Experimental, theoretical and docking studies of 2-hydroxy Schiff base type compounds derived from 2-amino-4-chlorobenzenethiol. J Mol Struct 2015. [DOI: 10.1016/j.molstruc.2015.08.026] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
297
|
Kesarwani M, Huber E, Kincaid Z, Evelyn CR, Biesiada J, Rance M, Thapa MB, Shah NP, Meller J, Zheng Y, Azam M. Targeting substrate-site in Jak2 kinase prevents emergence of genetic resistance. Sci Rep 2015; 5:14538. [PMID: 26419724 PMCID: PMC4588578 DOI: 10.1038/srep14538] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 08/18/2015] [Indexed: 12/13/2022] Open
Abstract
Emergence of genetic resistance against kinase inhibitors poses a great challenge for durable therapeutic response. Here, we report a novel mechanism of JAK2 kinase inhibition by fedratinib (TG101348) that prevents emergence of genetic resistance. Using in vitro drug screening, we identified 211 amino-acid substitutions conferring resistance to ruxolitinib (INCB018424) and cross-resistance to the JAK2 inhibitors AZD1480, CYT-387 and lestaurtinib. In contrast, these resistant variants were fully sensitive to fedratinib. Structural modeling, coupled with mutagenesis and biochemical studies, revealed dual binding sites for fedratinib. In vitro binding assays using purified proteins showed strong affinity for the substrate-binding site (Kd = 20 nM) while affinity for the ATP site was poor (Kd = ~8 μM). Our studies demonstrate that mutations affecting the substrate-binding pocket encode a catalytically incompetent kinase, thereby preventing emergence of resistant variants. Most importantly, our data suggest that in order to develop resistance-free kinase inhibitors, the next-generation drug design should target the substrate-binding site.
Collapse
Affiliation(s)
- Meenu Kesarwani
- Cincinnati Children's Hospital Medical Center, Cancer Blood Disease Institute, Divisions of Experimental Hematology and Cancer Pathology, Cincinnati, Ohio, 45229 USA
| | - Erika Huber
- Cincinnati Children's Hospital Medical Center, Cancer Blood Disease Institute, Divisions of Experimental Hematology and Cancer Pathology, Cincinnati, Ohio, 45229 USA
| | - Zachary Kincaid
- Cincinnati Children's Hospital Medical Center, Cancer Blood Disease Institute, Divisions of Experimental Hematology and Cancer Pathology, Cincinnati, Ohio, 45229 USA
| | - Chris R Evelyn
- Cincinnati Children's Hospital Medical Center, Cancer Blood Disease Institute, Divisions of Experimental Hematology and Cancer Pathology, Cincinnati, Ohio, 45229 USA
| | - Jacek Biesiada
- Cincinnati Children's Hospital Medical Center, Cancer Blood Disease Institute, Divisions of Experimental Hematology and Cancer Pathology, Cincinnati, Ohio, 45229 USA
| | - Mark Rance
- Department of Molecular Genetics, Biochemistry and Microbiology University of Cincinnati College of Medicine, University of Cincinnati, Ohio 45229 USA
| | - Mahendra B Thapa
- Department of Molecular Genetics, Biochemistry and Microbiology University of Cincinnati College of Medicine, University of Cincinnati, Ohio 45229 USA
| | - Neil P Shah
- Division of Hematology-Oncology UCSF School of Medicine, San Francisco, California, 94143 USA
| | - Jarek Meller
- Cincinnati Children's Hospital Medical Center, Cancer Blood Disease Institute, Divisions of Experimental Hematology and Cancer Pathology, Cincinnati, Ohio, 45229 USA
| | - Yi Zheng
- Cincinnati Children's Hospital Medical Center, Cancer Blood Disease Institute, Divisions of Experimental Hematology and Cancer Pathology, Cincinnati, Ohio, 45229 USA
| | - Mohammad Azam
- Cincinnati Children's Hospital Medical Center, Cancer Blood Disease Institute, Divisions of Experimental Hematology and Cancer Pathology, Cincinnati, Ohio, 45229 USA.,Department of Molecular Genetics, Biochemistry and Microbiology University of Cincinnati College of Medicine, University of Cincinnati, Ohio 45229 USA
| |
Collapse
|
298
|
O’Dwyer CA, O’Brien ME, Wormald MR, White MM, Banville N, Hurley K, McCarthy C, McElvaney NG, Reeves EP. The BLT1 Inhibitory Function of α-1 Antitrypsin Augmentation Therapy Disrupts Leukotriene B4Neutrophil Signaling. THE JOURNAL OF IMMUNOLOGY 2015; 195:3628-41. [DOI: 10.4049/jimmunol.1500038] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Accepted: 08/12/2015] [Indexed: 12/15/2022]
|
299
|
Hayden EY, Yamin G, Beroukhim S, Chen B, Kibalchenko M, Jiang L, Ho L, Wang J, Pasinetti GM, Teplow DB. Inhibiting amyloid β-protein assembly: Size-activity relationships among grape seed-derived polyphenols. J Neurochem 2015; 135:416-30. [PMID: 26228682 DOI: 10.1111/jnc.13270] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 07/22/2015] [Indexed: 12/25/2022]
Abstract
Epidemiological evidence that red wine consumption negatively correlates with risk of Alzheimer's disease has led to experimental studies demonstrating that grape seed extracts inhibit the aggregation and oligomerization of Aβ in vitro and ameliorate neuropathology and behavioral deficits in a mouse model of Alzheimer's disease. The active agent in the extracts is a mixed population of polyphenolic compounds. To evaluate the relative potency of each of these compounds, HPLC was used to fractionate the mixture into monomers, dimers, and oligomers. Each fraction was analyzed for its effect on Aβ conformational dynamics (circular dichroism), oligomerization (zero-length photochemical cross-linking), aggregation kinetics (Thioflavin T fluorescence), and morphology (electron microscopy). The relative activities of each fraction were determined on the basis of molar concentration (mol/L) or mass concentration (g/L). When molar concentration, the number concentration of each polyphenolic compound, was considered, the oligomer fraction was the most potent inhibitor of Aβ oligomerization and aggregation. However, when mass concentration, the number concentration of phenolic groups, was considered, monomers were the most potent inhibitors. To understand these ostensibly contradictory results, a model of polyphenol:Aβ complexation was developed. This model, which was found to be consistent with published X-ray crystallographic studies, offers an explanation for the effects of functional group polyvalency on inhibitor activity. Our data emphasize the importance of an in-depth understanding of the mechanism(s) underlying 'concentration dependence' in inhibitor systems involving polyfunctional agents.
Collapse
Affiliation(s)
- Eric Y Hayden
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Ghiam Yamin
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, California, USA.,Medical Scientist Training Program, Neuroscience Interdepartmental Ph.D. Program, University of California, Los Angeles, California, USA
| | - Shiela Beroukhim
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Benson Chen
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Mikhail Kibalchenko
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Lin Jiang
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Lap Ho
- Departments of Psychiatry and Neuroscience, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, New York, USA
| | - Jun Wang
- Departments of Psychiatry and Neuroscience, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, New York, USA.,Department of Neurology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, New York, USA.,Geriatric Research, Education and Clinical Center (GRECC), James J. Peters Veterans Affairs Medical Center, 130 West Kingsbridge Road, Bronx, New York, USA
| | - Giulio M Pasinetti
- Departments of Psychiatry and Neuroscience, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, New York, USA.,Department of Neurology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, New York, USA.,Geriatric Research, Education and Clinical Center (GRECC), James J. Peters Veterans Affairs Medical Center, 130 West Kingsbridge Road, Bronx, New York, USA
| | - David B Teplow
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, California, USA.,Molecular Biology Institute (MBI), and Brain Research Institute (BRI), David Geffen School of Medicine, University of California, Los Angeles, California, USA
| |
Collapse
|
300
|
Germane KL, Servinsky MD, Gerlach ES, Sund CJ, Hurley MM. Structural analysis of Clostridium acetobutylicum ATCC 824 glycoside hydrolase from CAZy family GH105. Acta Crystallogr F Struct Biol Commun 2015; 71:1100-8. [PMID: 26249707 PMCID: PMC4528949 DOI: 10.1107/s2053230x15012121] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 06/24/2015] [Indexed: 11/10/2022] Open
Abstract
Clostridium acetobutylicum ATCC 824 gene CA_C0359 encodes a putative unsaturated rhamnogalacturonyl hydrolase (URH) with distant amino-acid sequence homology to YteR of Bacillus subtilis strain 168. YteR, like other URHs, has core structural homology to unsaturated glucuronyl hydrolases, but hydrolyzes the unsaturated disaccharide derivative of rhamnogalacturonan I. The crystal structure of the recombinant CA_C0359 protein was solved to 1.6 Å resolution by molecular replacement using the phase information of the previously reported structure of YteR (PDB entry 1nc5) from Bacillus subtilis strain 168. The YteR-like protein is a six-α-hairpin barrel with two β-sheet strands and a small helix overlaying the end of the hairpins next to the active site. The protein has low primary protein sequence identity to YteR but is structurally similar. The two tertiary structures align with a root-mean-square deviation of 1.4 Å and contain a highly conserved active pocket. There is a conserved aspartic acid residue in both structures, which has been shown to be important for hydration of the C=C bond during the release of unsaturated galacturonic acid by YteR. A surface electrostatic potential comparison of CA_C0359 and proteins from CAZy families GH88 and GH105 reveals the make-up of the active site to be a combination of the unsaturated rhamnogalacturonyl hydrolase and the unsaturated glucuronyl hydrolase from Bacillus subtilis strain 168. Structural and electrostatic comparisons suggests that the protein may have a slightly different substrate specificity from that of YteR.
Collapse
Affiliation(s)
- Katherine L. Germane
- Oak Ridge Associated Universities, 4692 Millennium Drive, Suite 101, Belcamp, MD 21017, USA
| | - Matthew D. Servinsky
- RDRL-SEE-B, US Army Research Laboratory, 2800 Powder Mill Road, Adelphi, MD 20783, USA
| | - Elliot S. Gerlach
- Federal Staffing Resources, 2200 Somerville Road, Annapolis, MD 21401, USA
| | - Christian J. Sund
- RDRL-SEE-B, US Army Research Laboratory, 2800 Powder Mill Road, Adelphi, MD 20783, USA
| | - Margaret M. Hurley
- RDRL-SEE-B, US Army Research Laboratory, 4600 Deer Creek Loop, Aberdeen Proving Ground, MD 21005, USA
| |
Collapse
|