1
|
Olson KM, Devereaux AL, Chatterjee P, Saldaña-Shumaker SL, Shafer A, Plotkin A, Kandasamy R, MacKerell AD, Traynor JR, Cunningham CW. Nitro-benzylideneoxymorphone, a bifunctional mu and delta opioid receptor ligand with high mu opioid receptor efficacy. Front Pharmacol 2023; 14:1230053. [PMID: 37469877 PMCID: PMC10352325 DOI: 10.3389/fphar.2023.1230053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Accepted: 06/19/2023] [Indexed: 07/21/2023] Open
Abstract
Introduction: There is a major societal need for analgesics with less tolerance, dependence, and abuse liability. Preclinical rodent studies suggest that bifunctional ligands with both mu (MOPr) and delta (DOPr) opioid peptide receptor activity may produce analgesia with reduced tolerance and other side effects. This study explores the structure-activity relationships (SAR) of our previously reported MOPr/DOPr lead, benzylideneoxymorphone (BOM) with C7-methylene-substituted analogs. Methods: Analogs were synthesized and tested in vitro for opioid receptor binding and efficacy. One compound, nitro-BOM (NBOM, 12) was evaluated for antinociceptive effects in the warm water tail withdrawal assay in C57BL/6 mice. Acute and chronic antinociception was determined, as was toxicologic effects on chronic administration. Molecular modeling experiments were performed using the Site Identification by Ligand Competitive Saturation (SILCS) method. Results: NBOM was found to be a potent MOPr agonist/DOPr partial agonist that produces high-efficacy antinociception. Antinociceptive tolerance was observed, as was weight loss; this toxicity was only observed with NBOM and not with BOM. Modeling supports the hypothesis that the increased MOPr efficacy of NBOM is due to the substituted benzylidene ring occupying a nonpolar region within the MOPr agonist state. Discussion: Though antinociceptive tolerance and non-specific toxicity was observed on repeated administration, NBOM provides an important new tool for understanding MOPr/DOPr pharmacology.
Collapse
Affiliation(s)
- Keith M. Olson
- Department of Pharmacology and Edward F. Domino Research Center, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Andrea L. Devereaux
- Department of Pharmaceutical Sciences, Concordia University Wisconsin School of Pharmacy, Mequon, WI, United States
| | - Payal Chatterjee
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, United States
| | - Savanah L. Saldaña-Shumaker
- Department of Pharmaceutical Sciences, Concordia University Wisconsin School of Pharmacy, Mequon, WI, United States
| | - Amanda Shafer
- Department of Pharmacology and Edward F. Domino Research Center, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Adam Plotkin
- Department of Pharmaceutical Sciences, Concordia University Wisconsin School of Pharmacy, Mequon, WI, United States
| | - Ram Kandasamy
- Department of Pharmacology and Edward F. Domino Research Center, University of Michigan Medical School, Ann Arbor, MI, United States
- Department of Psychology, California State University, East Bay, Hayward, CA, United States
| | - Alexander D. MacKerell
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, United States
| | - John R. Traynor
- Department of Pharmacology and Edward F. Domino Research Center, University of Michigan Medical School, Ann Arbor, MI, United States
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, MI, United States
| | - Christopher W. Cunningham
- Department of Pharmaceutical Sciences, Concordia University Wisconsin School of Pharmacy, Mequon, WI, United States
| |
Collapse
|
2
|
Ganai I, Saha I, Banerjee P, Laha A, Sultana S, Sultana N, Biswas H, Moitra S, Podder S. In silico analysis of single nucleotide polymorphism (rs34377097) of TBXA2R gene and pollen induced bronchial asthma susceptibility in West Bengal population, India. Front Immunol 2023; 14:1089514. [PMID: 36936944 PMCID: PMC10018227 DOI: 10.3389/fimmu.2023.1089514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 02/14/2023] [Indexed: 03/06/2023] Open
Abstract
Introduction Prevalence of asthma is increasing steadily among general population in developing countries over past two decades. One of the causative agents of broncho-constriction in asthma is thromboxane A2 receptor (TBXA2R). However few studies of TBXA2R polymorphism were performed so far. The present study aimed to assess potential association of TBXA2R rs34377097 polymorphism causing missense substitution of Arginine to Leucine (R60L) among 482 patients diagnosed with pollen-induced asthma and 122 control participants from West Bengal, India. Also we performed in-silico analysis of mutated TBXA2R protein (R60L) using homology modeling. Methods Clinical parameters like Forced expiratory volume in 1 second (FEV1), FEV1/Forced vital capacity (FVC) and Peak expiratory flow rate (PEFR) were assessed using spirometry. Patients' sensitivity was measured by skin prick test (SPT) against 16 pollen allergens. Polymerase chain reaction-based Restriction fragment length polymorphism was done for genotyping. Structural model of wild type and homology model of polymorphic TBXA2R was generated using AlphaFold2 and MODELLER respectively. Electrostatic surface potential was calculated using APBS plugin in PyMol. Results Genotype frequencies differed significantly between the study groups (P=0.03). There was no significant deviation from Hardy-Weinberg equilibrium in control population (χ2=1.56). Asthmatic patients have significantly higher frequency of rs34377097TT genotype than control subjects (P=0.03). SPT of patients showed maximum sensitivity in A. indica (87.68%) followed by C. nusifera (83.29%) and C. pulcherima (74.94%). Significant difference existed for pollen sensitivity in adolescent and young adult (P=0.01) and between young and old adult (P=0.0003). Significant negative correlation was found between FEV1/FVC ratio and intensity of SPT reactions (P<0.0001). Significant association of FEV1, FEV1/FVC and PEFR was observed with pollen-induced asthma. Furthermore, risk allele T was found to be clinically correlated with lower FEV1/FVC ratio (P=0.015) in patients. Our data showed R60L polymorphism, which was conserved across mammals, significantly reduced positive electrostatic charge of polymorphic protein in cytoplasmic domain thus altered downstream pathway and induced asthma response. Discussion The present in-silico study is the first one to report association of TBXA2R rs34377097 polymorphism in an Indian population. It may be used as prognostic marker of clinical response to asthma in West Bengal and possible target of therapeutics in future.
Collapse
Affiliation(s)
- Indranil Ganai
- Ecology and Allergology Lab, Department of Zoology, The University of Burdwan, Burdwan, India
| | - Ishita Saha
- Department of Physiology, Medical College and Hospital, Kolkata, India
| | - Priyajit Banerjee
- Fishery and Eco-toxicology Research Lab, Department of Zoology, The University of Burdwan, Burdwan, India
| | - Arghya Laha
- Ecology and Allergology Lab, Department of Zoology, The University of Burdwan, Burdwan, India
| | - Saheen Sultana
- Ecology and Allergology Lab, Department of Zoology, The University of Burdwan, Burdwan, India
| | - Nasima Sultana
- Ecology and Allergology Lab, Department of Zoology, The University of Burdwan, Burdwan, India
| | - Himani Biswas
- Post Graduate Department of Zoology, Krishnagar Government College, Krishnagar, India
| | | | - Sanjoy Podder
- Ecology and Allergology Lab, Department of Zoology, The University of Burdwan, Burdwan, India
- *Correspondence: Sanjoy Podder,
| |
Collapse
|
3
|
Garai P, Banerjee P, Sharma P, Chatterjee A, Bhattacharya R, Saha NC. Mechanistic insights to lactic and formic acid toxicity on benthic oligochaete worm Tubifex tubifex. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:87319-87333. [PMID: 35802337 DOI: 10.1007/s11356-022-21361-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 06/05/2022] [Indexed: 06/15/2023]
Abstract
Lactic and formic acid are two commonly found monocarboxylic organic acids. Lactic acid is discharged into the water bodies as acidic industrial effluent from the food, cosmetic, chemical, and pharmaceutical industries, whereas formic acid is discharged from various paper, leather tanning, and textile processing industries. The present study investigated the toxicity of both organic acids upon the benthic oligochaete worm Tubifex tubifex. The 96-h median lethal concentration (LC50) values for lactic and formic acid are determined as 143.81 mg/l and 57.99 mg/l respectively. The effects of two sublethal concentrations (10% and 30% of 96 h LC50) of these acids on differential expression of oxidative stress enzymes are investigated. The comparative analysis of acute toxicity demonstrates that formic acid exposure is more detrimental to T. tubifex than lactic acid. The in silico structural analysis predicts that formic acid can interact with cytochrome c oxidase of the electron transport system and thereby inhibits its functionality and induces reactive oxygen species production. Integrated biomarker response (IBR) analysis illustrates that overall oxidative stress of formic acid to T. tubifex is significantly higher than that of lactic acid, which supports the structural analysis. It is concluded from this study that toxicokinetic-toxicodynamic and species sensitivity distributions studies are helpful for ecological risk management of environmental toxicants.
Collapse
Affiliation(s)
- Pramita Garai
- Fisheries and Ecotoxicology Research Laboratory (Vice-Chancellor's Research Group), Department of Zoology, The University of Burdwan, Burdwan, West Bengal, 713104, India
| | - Priyajit Banerjee
- Fisheries and Ecotoxicology Research Laboratory (Vice-Chancellor's Research Group), Department of Zoology, The University of Burdwan, Burdwan, West Bengal, 713104, India
| | - Pramita Sharma
- Fisheries and Ecotoxicology Research Laboratory (Vice-Chancellor's Research Group), Department of Zoology, The University of Burdwan, Burdwan, West Bengal, 713104, India
| | - Arnab Chatterjee
- Fisheries and Ecotoxicology Research Laboratory (Vice-Chancellor's Research Group), Department of Zoology, The University of Burdwan, Burdwan, West Bengal, 713104, India
| | - Ritwick Bhattacharya
- Fisheries and Ecotoxicology Research Laboratory (Vice-Chancellor's Research Group), Department of Zoology, The University of Burdwan, Burdwan, West Bengal, 713104, India
| | - Nimai Chandra Saha
- Fisheries and Ecotoxicology Research Laboratory (Vice-Chancellor's Research Group), Department of Zoology, The University of Burdwan, Burdwan, West Bengal, 713104, India.
| |
Collapse
|
4
|
Lummis SCR, Dougherty DA. Expression of Mutant Glycine Receptors in Xenopus Oocytes Using Canonical and Non-Canonical Amino Acids Reveals Distinct Roles of Conserved Proline Residues. MEMBRANES 2022; 12:1012. [PMID: 36295771 PMCID: PMC9607081 DOI: 10.3390/membranes12101012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/05/2022] [Accepted: 10/08/2022] [Indexed: 06/16/2023]
Abstract
Pentameric ligand-gated ion channels (pLGIC) play important roles in fast neuronal signal transmission. Functional receptors are pentamers, with each subunit having an extracellular domain (ECD), a transmembrane domain (TMD) and an intracellular domain. The binding of the agonist to the ECD induces a structural change that is transduced to the TMD to open the channel. Molecular details of this process are emerging, but a comprehensive understanding is still lacking. Proline (Pro) is one amino acid that has attracted much interest; its unusual features generate bends in loops and kinks and bulges in helices, which can be essential for function in some pLGICs. Here, we explore the roles of four conserved Pros in the glycine receptor (GlyR), creating substitutions with canonical and noncanonical amino acids, characterizing them using two electrode voltage clamp electrophysiology in Xenopus oocytes, and interpreting changes in receptor parameters using structural data from the open and closed states of the receptor. The data reveal that for efficient function, the Pro in the α1β1 loop is needed to create a turn and to be the correct size and shape to interact with nearby residues; the peptide bond of the Pro in the Cys-loop requires the cis conformation; and the Pros in loop A and M1 allow efficient function because of their reduced hydrogen bonding capacity. These data are broadly consistent with data from other pLGICs, and therefore likely represent the important features of these Pros in all members of the family.
Collapse
Affiliation(s)
- Sarah C. R. Lummis
- Department of Biochemistry, University of Cambridge, Tennis Court Road, Cambridge CB2 1GA, UK
| | - Dennis A. Dougherty
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| |
Collapse
|
5
|
Force Field Parameters for Fe 2+4S 2-4 Clusters of Dihydropyrimidine Dehydrogenase, the 5-Fluorouracil Cancer Drug Deactivation Protein: A Step towards In Silico Pharmacogenomics Studies. Molecules 2021; 26:molecules26102929. [PMID: 34069161 PMCID: PMC8156676 DOI: 10.3390/molecules26102929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/07/2021] [Accepted: 05/11/2021] [Indexed: 12/01/2022] Open
Abstract
The dimeric dihydropyrimidine dehydrogenase (DPD), metalloenzyme, an adjunct anti-cancer drug target, contains highly specialized 4 × Fe2+4S2−4 clusters per chain. These clusters facilitate the catalysis of the rate-limiting step in the pyrimidine degradation pathway through a harmonized electron transfer cascade that triggers a redox catabolic reaction. In the process, the bulk of the administered 5-fluorouracil (5-FU) cancer drug is inactivated, while a small proportion is activated to nucleic acid antimetabolites. The occurrence of missense mutations in DPD protein within the general population, including those of African descent, has adverse toxicity effects due to altered 5-FU metabolism. Thus, deciphering mutation effects on protein structure and function is vital, especially for precision medicine purposes. We previously proposed combining molecular dynamics (MD) and dynamic residue network (DRN) analysis to decipher the molecular mechanisms of missense mutations in other proteins. However, the presence of Fe2+4S2−4 clusters in DPD poses a challenge for such in silico studies. The existing AMBER force field parameters cannot accurately describe the Fe2+ center coordination exhibited by this enzyme. Therefore, this study aimed to derive AMBER force field parameters for DPD enzyme Fe2+ centers, using the original Seminario method and the collation features Visual Force Field Derivation Toolkit as a supportive approach. All-atom MD simulations were performed to validate the results. Both approaches generated similar force field parameters, which accurately described the human DPD protein Fe2+4S2−4 cluster architecture. This information is crucial and opens new avenues for in silico cancer pharmacogenomics and drug discovery related research on 5-FU drug efficacy and toxicity issues.
Collapse
|
6
|
Gezdirici A, Teralı K, Gülec EY, Bornaun H, Dogan M, Eröz R. An integrated clinical and molecular study of a cohort of Turkish patients with Marfan syndrome harboring known and novel FBN1 variants. J Hum Genet 2021; 66:647-657. [PMID: 33483584 DOI: 10.1038/s10038-021-00899-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 12/12/2020] [Accepted: 01/04/2021] [Indexed: 11/09/2022]
Abstract
Marfan syndrome (MFS) is an autosomal dominant genetic condition that mainly affects connective tissue in many parts of the body. Cardinal manifestations involve the ocular, skeletal, and cardiovascular systems. The diagnosis of MFS relies on the revised Ghent criteria, outlined by international expert opinion to facilitate accurate recognition of this syndrome as well as to improve patient management and counseling. However, it may not always be possible to make a definitive diagnosis according to these criteria in each patient and thus molecular confirmation is necessary in subjects with suspected MFS. This debilitating, if not fatal, disorder is caused by mutations in FBN1, which encodes a major constitutive element of extracellular microfibrils. Here, we present a detailed clinical and molecular analysis of 76 Turkish patients with definitive or suspected MFS diagnosed at our center between 2014 and 2019. We were able to identify a total of 51 different FBN1 variants in our cohort, 31 of which have previously been reported in the relevant scientific literature. The remaining 20 variants have not been documented to date. In one patient, we detected a large deletion including the entire FBN1 gene using the array CGH approach. Currently, there are very few studies on the genotype-phenotype correlation of patients with MFS, and no clear genotype-phenotype maps for MFS have been constructed so far, except for some cases. We believe that our findings will make a rich and peculiar contribution to the elusive genotype-phenotype relationship in MFS, especially in this large and populous ethnic group.
Collapse
Affiliation(s)
- Alper Gezdirici
- Department of Medical Genetics, Basaksehir Cam and Sakura City Hospital, 34480, Istanbul, Turkey.
| | - Kerem Teralı
- Department of Medical Biochemistry, Faculty of Medicine, Near East University, 99138, Nicosia, Cyprus.,Bioinformatics & Computational Biology Research Group, DESAM Institute, Near East University, 99138, Nicosia, Cyprus
| | - Elif Yılmaz Gülec
- Department of Medical Genetics, Kanuni Sultan Suleyman Training and Research Hospital, 34303, Istanbul, Turkey
| | - Helen Bornaun
- Department of Pediatric Cardiology, Kanuni Sultan Suleyman Training and Research Hospital, 34303, Istanbul, Turkey
| | - Mustafa Dogan
- Department of Medical Genetics, Faculty of Medicine, Malatya Turgut Ozal University, 44300, Malatya, Turkey
| | - Recep Eröz
- Department of Medical Genetics, Faculty of Medicine, Duzce University, 81010, Düzce, Turkey
| |
Collapse
|
7
|
El Jahrani N, Cretin G, de Brevern AG. CALR-ETdb, the database of calreticulin variants diversity in essential thrombocythemia. Platelets 2021; 33:157-167. [PMID: 33444113 DOI: 10.1080/09537104.2020.1869712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Essential thrombocythemia (ET) is a blood cancer defined by a strong increase of platelet numbers. A quarter of patients suffering from ET show mutations in the last exon of calreticulin (CALR) gene. Two variants named type 1 and type 2 represent 85% of these patients. However, a large number of other variants have been determined. In this study, we have compiled variants taken from COSMIC database and literature leading to 155 different variants. This large number of variants allowed redefining 5 new classes extending the classification of type 1-like and type 2-like to a finer description. These analyses showed that last class, named E, corresponding to more than 10% of CALR variants seemed not attached to ET. Structural properties analyzed showed that CALR variants associated to ET have common features. All the compiled and refined information had been included into a freely dedicated database CALR-ETdb (https://www.dsimb.inserm.fr/CALR-ET).
Collapse
Affiliation(s)
- Nora El Jahrani
- Université de Paris, UMR_S 1134, Université De La Réunion, Université Des Antilles, Paris, France.,INSERM, U 1134, DSIMB, Paris, France.,Institut National De La Transfusion Sanguine (INTS), Paris, France.,Laboratoire d'Excellence GR-Ex, Paris, France
| | - Gabriel Cretin
- Université de Paris, UMR_S 1134, Université De La Réunion, Université Des Antilles, Paris, France.,INSERM, U 1134, DSIMB, Paris, France.,Institut National De La Transfusion Sanguine (INTS), Paris, France.,Laboratoire d'Excellence GR-Ex, Paris, France
| | - Alexandre G de Brevern
- Université de Paris, UMR_S 1134, Université De La Réunion, Université Des Antilles, Paris, France.,INSERM, U 1134, DSIMB, Paris, France.,Institut National De La Transfusion Sanguine (INTS), Paris, France.,Laboratoire d'Excellence GR-Ex, Paris, France
| |
Collapse
|
8
|
Abstract
Genome sequencing projects have resulted in a rapid increase in the number of known protein sequences. In contrast, only about one-hundredth of these sequences have been characterized at atomic resolution using experimental structure determination methods. Computational protein structure modeling techniques have the potential to bridge this sequence-structure gap. In the following chapter, we present an example that illustrates the use of MODELLER to construct a comparative model for a protein with unknown structure. Automation of a similar protocol has resulted in models of useful accuracy for domains in more than half of all known protein sequences.
Collapse
|
9
|
Baker BW, Dougherty DA, Lummis SCR. Proline Residues Contribute to Efficient GABA p Receptor Function. ACS Chem Neurosci 2020; 11:4215-4222. [PMID: 33201686 DOI: 10.1021/acschemneuro.0c00483] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
GABAp receptors are homomeric pentameric ligand-gated ion channels (pLGICs) and are useful for probing the molecular details of the mechanism of action in this important protein family. Here, we explore the role of proline (Pro) residues by creating mutant receptors, expressing them in HEK293 cells, and using fluorescent membrane potential sensitive dye to monitor receptor activity. The data revealed that 3 of the Pro-to-alanine substitutions resulted in nonfunctional receptors (one in the Cys-loop, one in loop A and one in the β2-β3 loop), 7 resulted in increased EC50 values, and the remaining 13 resulted in receptors with properties similar to WT receptors. Further exploration of the nonfunctional mutants using expression in Xenopus laevis oocytes and whole-cell voltage-clamp electrophysiology, incorporating both canonical and noncanonical amino acids, revealed that the Pro in the Cys-loop had a preference for analogues with a high intrinsic cis bias, the Pro in loop A required a ring, and the β2-β3 loop Pro contributes to expression. The data from the Cys-loop Pro are consistent with those from other pLGICs, while those of the loop A Pro and some of the other Pros surprisingly differ. Thus, overall, the data clarify the roles of many of the Pros in the GABAp receptor and also suggest that caution must be applied in using data from one receptor to understand molecular details of all pLGICs.
Collapse
Affiliation(s)
- Benjamin W. Baker
- Department of Biochemistry, University of Cambridge, Tennis Court Road, Cambridge CB2 1GA, United Kingdom
| | - Dennis A. Dougherty
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, California 91125, United States
| | - Sarah C. R. Lummis
- Department of Biochemistry, University of Cambridge, Tennis Court Road, Cambridge CB2 1GA, United Kingdom
| |
Collapse
|
10
|
Hughes ME, Mesoy SM, Capes E, Lummis SCR. Many Proline Residues in the Extracellular Domain Contribute to Glycine Receptor Function. ACS Chem Neurosci 2020; 11:2658-2665. [PMID: 32786326 DOI: 10.1021/acschemneuro.0c00320] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Prolines in signaling proteins are of particular interest because they have a range of unique properties that may be critical for function. Here we show that many proline residues in the extracellular domain (ECD) of the glycine receptor are involved in the correct functioning of this ligand-gated ion channel. We explore their role by creating mutant receptors, expressing them in cells, and using fluorescent membrane potential sensitive dye to monitor receptor activity. We then interpret the changes in receptor parameters using structural information from the open and closed states of the receptor. The data reveal that substitution with alanine of ten of the 13 Pro residues in the ECD alters the function of the receptor: one substitution ablates function, six cause a decrease in the EC50, and three cause an increase. Only three of these mutants result in EC50 values similar to WT. The nonfunctional mutant, Pro30Ala, was further probed in oocytes, and the data suggest a role in both expression and function. Examination of the locations of sensitive Pro residues in the receptor and identification of potential interactions with nearby residues reveal how these residues could contribute to the correct functioning of this typical pentameric ligand-gated ion channel.
Collapse
Affiliation(s)
- Merryn E. Hughes
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1QW, U.K
| | - Susanne M. Mesoy
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1QW, U.K
| | - Emily Capes
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1QW, U.K
| | - Sarah C. R. Lummis
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1QW, U.K
| |
Collapse
|
11
|
Dohrke JN, Watson JF, Birchall K, Greger IH. Characterizing the binding and function of TARP γ8-selective AMPA receptor modulators. J Biol Chem 2020; 295:14565-14577. [PMID: 32747446 PMCID: PMC7586208 DOI: 10.1074/jbc.ra120.014135] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 07/31/2020] [Indexed: 12/21/2022] Open
Abstract
α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid(AMPA)-type glutamate receptors (AMPARs) are the predominant excitatory neurotransmitter receptors in the brain, where they mediate synaptic transmission and plasticity. Excessive AMPAR activation leads to diseases such as epilepsy. AMPAR properties are modulated by auxiliary proteins and foremost by the transmembrane AMPAR regulatory proteins (TARPs). These distribute in unique expression patterns across the brain, rendering AMPAR/TARP complexes promising targets for region-specific therapeutic intervention. TARP γ8 is predominantly expressed in the forebrain and is enriched in the hippocampus, a region associated with temporal lobe epilepsy. Recent high-throughput medicinal chemistry screens have identified multiple promising compounds that selectively target AMPARs associated with γ8 and hold promise for epilepsy treatment. However, how these modulators target the receptor complex is currently unknown. Here, we use a combination of ligand docking, molecular dynamics simulations, and electrophysiology to address this question. We identify a conserved oxindole isostere, shared between three structurally diverse modulators (LY-3130481, JNJ-55511118, and JNJ-61432059) as the major module engaging γ8 by an H-bond to Asn-172 (γ8). The remaining variable region of each molecule likely targets the receptor complex in ligand-selective modes. Functional data reveal parallels in the underlying modulatory action of two prominent compounds. This work will aid development of refined AMPAR epilepsy therapeutics and facilitate to uncover the mechanisms by which TARPs modulate the receptor.
Collapse
Affiliation(s)
- Jan-Niklas Dohrke
- Neurobiology Division, Medical Research Council Laboratory of Molecular Biology, Cambridge, United Kingdom
| | - Jake F Watson
- Neurobiology Division, Medical Research Council Laboratory of Molecular Biology, Cambridge, United Kingdom
| | | | - Ingo H Greger
- Neurobiology Division, Medical Research Council Laboratory of Molecular Biology, Cambridge, United Kingdom.
| |
Collapse
|
12
|
Mariano D, Pantuza N, Santos LH, Rocha REO, de Lima LHF, Bleicher L, de Melo-Minardi RC. Glutantβase: a database for improving the rational design of glucose-tolerant β-glucosidases. BMC Mol Cell Biol 2020; 21:50. [PMID: 32611314 PMCID: PMC7329481 DOI: 10.1186/s12860-020-00293-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 06/22/2020] [Indexed: 11/22/2022] Open
Abstract
Β-glucosidases are key enzymes used in second-generation biofuel production. They act in the last step of the lignocellulose saccharification, converting cellobiose in glucose. However, most of the β-glucosidases are inhibited by high glucose concentrations, which turns it a limiting step for industrial production. Thus, β-glucosidases have been targeted by several studies aiming to understand the mechanism of glucose tolerance, pH and thermal resistance for constructing more efficient enzymes. In this paper, we present a database of β-glucosidase structures, called Glutantβase. Our database includes 3842 GH1 β-glucosidase sequences collected from UniProt. We modeled the sequences by comparison and predicted important features in the 3D-structure of each enzyme. Glutantβase provides information about catalytic and conserved amino acids, residues of the coevolution network, protein secondary structure, and residues located in the channel that guides to the active site. We also analyzed the impact of beneficial mutations reported in the literature, predicted in analogous positions, for similar enzymes. We suggested these mutations based on six previously described mutants that showed high catalytic activity, glucose tolerance, or thermostability (A404V, E96K, H184F, H228T, L441F, and V174C). Then, we used molecular docking to verify the impact of the suggested mutations in the affinity of protein and ligands (substrate and product). Our results suggest that only mutations based on the H228T mutant can reduce the affinity for glucose (product) and increase affinity for cellobiose (substrate), which indicates an increment in the resistance to product inhibition and agrees with computational and experimental results previously reported in the literature. More resistant β-glucosidases are essential to saccharification in industrial applications. However, thermostable and glucose-tolerant β-glucosidases are rare, and their glucose tolerance mechanisms appear to be related to multiple and complex factors. We gather here, a set of information, and made predictions aiming to provide a tool for supporting the rational design of more efficient β-glucosidases. We hope that Glutantβase can help improve second-generation biofuel production. Glutantβase is available at http://bioinfo.dcc.ufmg.br/glutantbase .
Collapse
Affiliation(s)
- Diego Mariano
- Laboratory of Bioinformatics and Systems. Department of Computer Science, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil.
| | - Naiara Pantuza
- Laboratory of Bioinformatics and Systems. Department of Computer Science, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - Lucianna H Santos
- Laboratory of Bioinformatics and Systems. Department of Computer Science, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - Rafael E O Rocha
- Laboratory of Bioinformatics and Systems. Department of Computer Science, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - Leonardo H F de Lima
- Laboratory of Molecular Modelling and Bioinformatics (LAMMB), Department of Physical and Biological Sciences, Universidade Federal de São João Del-Rei, Campus Sete Lagoas, Sete Lagoas, 35701-970, Brazil
| | - Lucas Bleicher
- Protein Computational Biology Laboratory, Department of Biochemistry and Immunology, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - Raquel Cardoso de Melo-Minardi
- Laboratory of Bioinformatics and Systems. Department of Computer Science, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil.
| |
Collapse
|
13
|
Ali A, Kumar R, Khan A, Khan AU. Interaction of LysM BON family protein domain with carbapenems: A putative mechanism of carbapenem resistance. Int J Biol Macromol 2020; 160:212-223. [PMID: 32464197 DOI: 10.1016/j.ijbiomac.2020.05.172] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 05/21/2020] [Accepted: 05/21/2020] [Indexed: 10/24/2022]
Abstract
Carbapenem resistance in Gram-negative pathogens has become a global concern for health workers worldwide. In one of our earlier studies, a Klebsiella pneumoniae-carbapenemase-2 producing strain was induced with meropenem to explore differentially expressed proteins under induced and uninduced conditions. There is, LysM domain BON family protein, was found over 12-fold expressed under the induced state. A hypothesis was proposed that LysM domain protein might have an affinity towards carbapenem antibiotics making them unavailable to bind with their target. Hence, we initiated a study to understand the binding mode of carbapenem with LysM domain protein. MICs of imipenem and meropenem against LysM clone were increased by several folds as compared to NP-6 clinical strain as well as DH5 α (PET-28a KPC-2) clone. This study further revealed a strong binding of both antibiotics to LysM domain protein. Molecular simulation studies of LysM domain protein with meropenem and imipenem for 80 ns has also showed stable structure. We concluded that overexpressed LysM domain under induced condition interacted with carbapenems, leading to enhanced resistance as proved by high MIC values. Hence, the study proved the proposed hypothesis that the LysM domain plays a significant role in the putative mechanism of antibiotics resistance.
Collapse
Affiliation(s)
- Abid Ali
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, UP, India
| | - Rakesh Kumar
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, Delhi 110067, India
| | - Arbab Khan
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, UP, India
| | - Asad U Khan
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, UP, India; Faculty of Science and Marine Environment, University Malysia Terengganu, Kuala Terengganu, Malaysia.
| |
Collapse
|
14
|
Zhu Z, Yi F, Epplin MP, Liu D, Summer SL, Mizu R, Shaulsky G, XiangWei W, Tang W, Burger PB, Menaldino DS, Myers SJ, Liotta DC, Hansen KB, Yuan H, Traynelis SF. Negative allosteric modulation of GluN1/GluN3 NMDA receptors. Neuropharmacology 2020; 176:108117. [PMID: 32389749 DOI: 10.1016/j.neuropharm.2020.108117] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 04/14/2020] [Accepted: 04/27/2020] [Indexed: 12/15/2022]
Abstract
NMDA receptors are ligand-gated ion channels that mediate excitatory neurotransmission. Most native NMDA receptors are tetrameric assemblies of two glycine-binding GluN1 and two glutamate-binding GluN2 subunits. Co-assembly of the glycine-binding GluN1 with glycine-binding GluN3 subunits (GluN3A-B) creates glycine activated receptors that possess strikingly different functional and pharmacological properties compared to GluN1/GluN2 NMDA receptors. The role of GluN1/GluN3 receptors in neuronal function remains unknown, in part due to lack of pharmacological tools with which to explore their physiological roles. We have identified the negative allosteric modulator EU1180-438, which is selective for GluN1/GluN3 receptors over GluN1/GluN2 NMDA receptors, AMPA, and kainate receptors. EU1180-438 is also inactive at GABA, glycine, and P2X receptors, but displays inhibition of some nicotinic acetylcholine receptors. Furthermore, we demonstrate that EU1180-438 produces robust inhibition of glycine-activated current responses mediated by native GluN1/GluN3A receptors in hippocampal CA1 pyramidal neurons. EU1180-438 is a non-competitive antagonist with activity that is independent of membrane potential (i.e. voltage-independent), glycine concentration, and extracellular pH. Non-stationary fluctuation analysis of neuronal current responses provided an estimated weighted mean unitary conductance of 6.1 pS for GluN1/GluN3A channels, and showed that EU1180-438 has no effect on conductance. Site-directed mutagenesis suggests that structural determinants of EU1180-438 activity reside near a short pre-M1 helix that lies parallel to the plane of the membrane below the agonist binding domain. These findings demonstrate that structural differences between GluN3 and other glutamate receptor subunits can be exploited to generate subunit-selective ligands with utility in exploring the roles GluN3 in neuronal function.
Collapse
Affiliation(s)
- Zongjian Zhu
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA; Department of Neonatology, First Affiliated Hospital of Xi'an Jiaotong University, 710061, Xi'an, Shaanxi, China
| | - Feng Yi
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT, 59812, USA
| | - Matthew P Epplin
- Department of Chemistry, Emory University, Atlanta, GA, 30322, USA
| | - Ding Liu
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | | | - Ruth Mizu
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Gil Shaulsky
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Wenshu XiangWei
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Weiting Tang
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Pieter B Burger
- Department of Chemistry, Emory University, Atlanta, GA, 30322, USA
| | | | - Scott J Myers
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Dennis C Liotta
- Department of Chemistry, Emory University, Atlanta, GA, 30322, USA
| | - Kasper B Hansen
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT, 59812, USA
| | - Hongjie Yuan
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Stephen F Traynelis
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA.
| |
Collapse
|
15
|
Loxoscelism: Advances and Challenges in the Design of Antibody Fragments with Therapeutic Potential. Toxins (Basel) 2020; 12:toxins12040256. [PMID: 32316084 PMCID: PMC7232456 DOI: 10.3390/toxins12040256] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 04/12/2020] [Accepted: 04/13/2020] [Indexed: 01/08/2023] Open
Abstract
Envenoming due to Loxosceles spider bites still remains a neglected disease of particular medical concern in the Americas. To date, there is no consensus for the treatment of envenomed patients, yet horse polyclonal antivenoms are usually infused to patients with identified severe medical conditions. It is widely known that venom proteins in the 30–35 kDa range with sphingomyelinase D (SMasesD) activity, reproduce most of the toxic effects observed in loxoscelism. Hence, we believe that monoclonal antibody fragments targeting such toxins might pose an alternative safe and effective treatment. In the present study, starting from the monoclonal antibody LimAb7, previously shown to target SMasesD from the venom of L. intermedia and neutralize its dermonecrotic activity, we designed humanized antibody V-domains, then produced and purified as recombinant single-chain antibody fragments (scFvs). These molecules were characterized in terms of humanness, structural stability, antigen-binding activity, and venom-neutralizing potential. Throughout this process, we identified some blocking points that can impact the Abs antigen-binding activity and neutralizing capacity. In silico analysis of the antigen/antibody amino acid interactions also contributed to a better understanding of the antibody’s neutralization mechanism and led to reformatting the humanized antibody fragment which, ultimately, recovered the functional characteristics for efficient in vitro venom neutralization.
Collapse
|
16
|
Study of alkaline phosphatase interaction with putrescine using multi-spectroscopic and docking methods. Colloids Surf B Biointerfaces 2020; 185:110509. [DOI: 10.1016/j.colsurfb.2019.110509] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 09/13/2019] [Accepted: 09/14/2019] [Indexed: 12/19/2022]
|
17
|
Costanzi S, Cohen A, Danfora A, Dolatmoradi M. Influence of the Structural Accuracy of Homology Models on Their Applicability to Docking-Based Virtual Screening: The β 2 Adrenergic Receptor as a Case Study. J Chem Inf Model 2019; 59:3177-3190. [PMID: 31257873 DOI: 10.1021/acs.jcim.9b00380] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
How accurate do structures of the β2 adrenergic receptor (β2AR) need to be to effectively serve as platforms for docking-based virtual screening campaigns? To answer this research question, here, we targeted through controlled virtual screening experiments 23 homology models of the β2AR endowed with different levels of structural accuracy. Subsequently, we studied the correlation between virtual screening performance and structural accuracy of the targeted models. Moreover, we studied the correlation between virtual screening performance and template/target receptor sequence identity. Our study demonstrates that docking-based virtual screening campaigns targeting homology models of the β2AR, in the majority of the cases, yielded results that exceeded random expectations in terms of area under the receiver operating characteristic curve (ROC AUC). Moreover, with the most effective scoring method, over one-third and one-quarter of the models yielded results that exceeded random expectation also in terms of enrichment factors (EF1, EF5, and EF10) and BEDROC (α = 160.9), respectively. Not surprisingly, we found a detectable linear correlation between virtual screening performance and structural accuracy of the ligand-binding cavity. We also found a detectable linear correlation between virtual screening performance and structural accuracy of the second extracellular loop (EL2). Finally, our data indicate that, although there is no detectable linear correlation between virtual screening performance and template/β2AR sequence identity, models built on the basis of templates that show high sequence identity with the β2AR, especially within the ligand-biding cavity, performed consistently well. Conversely, models with lower sequence identity displayed performance levels that ranged from very good to random, with no apparent correlation with the sequence identity itself.
Collapse
|
18
|
Investigating targets for neuropharmacological intervention by molecular dynamics simulations. Biochem Soc Trans 2019; 47:909-918. [PMID: 31085614 DOI: 10.1042/bst20190048] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 04/11/2019] [Accepted: 04/23/2019] [Indexed: 01/09/2023]
Abstract
Medical research has identified over 500 brain disorders. Among these, there are still only very few neuropathologies whose causes are fully understood and, consequently, very few drugs whose mechanism of action is known. No FDA drug has been identified for major neurodegenerative diseases, such as Alzheimer's and Parkinson's. We still lack effective treatments and strategies for modulating progression or even early neurodegenerative disease onset diagnostic tools. A great support toward the highly needed identification of neuroactive drugs comes from computer simulation methods and, in particular, from molecular dynamics (MD). This provides insight into structure-function relationship of a target and predicts structure, dynamics and energetics of ligand/target complexes under biologically relevant conditions like temperature and physiological saline concentration. Here, we present examples of the predictive power of MD for neuroactive ligands/target complexes. This brief survey from our own research shows the usefulness of partnerships between academia and industry, and from joint efforts between experimental and theoretical groups.
Collapse
|
19
|
Latif R, Mezei M, Morshed SA, Ma R, Ehrlich R, Davies TF. A Modifying Autoantigen in Graves' Disease. Endocrinology 2019; 160:1008-1020. [PMID: 30822352 PMCID: PMC6455603 DOI: 10.1210/en.2018-01048] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 02/26/2019] [Indexed: 12/22/2022]
Abstract
The TSH receptor (TSHR) is the major autoantigen in Graves' disease (GD). Bioinformatic analyses predict the existence of several human TSHR isoforms from alternative splicing, which can lead to the coexpression of multiple receptor forms. The most abundant of these is TSHRv1.3. In silico modeling of TSHRv1.3 demonstrated the structural integrity of this truncated receptor isoform and its potential binding of TSH. Tissue profiling revealed wide expression of TSHRv1.3, with a predominant presence in thyroid, bone marrow, thymus, and adipose tissue. To gain insight into the role of this v1.3 receptor isoform in thyroid pathophysiology, we cloned the entire open reading frame into a mammalian expression vector. Immunoprecipitation studies demonstrated that both TSHR-stimulating antibody and human TSH could bind v1.3. Furthermore, TSHRv1.3 inhibited the stimulatory effect of TSH and TSHR-Ab MS-1 antibody on TSHR-induced cAMP generation in a dose-dependent manner. To confirm the antigenicity of v1.3, we used a peptide ELISA against two different epitopes. Of 13 GD samples, 11 (84.6%) were positive for a carboxy terminal peptide and 10 (76.9%) were positive with a junction region peptide. To demonstrate that intracellular v1.3 could serve as an autoantigen and modulate disease, we used double-transfected Chinese hamster ovary cells that expressed both green fluorescent protein (GFP)-tagged TSHRv1.3 and full-length TSHR. We then induced cell stress and apoptosis using a TSHR monoclonal antibody and observed the culture supernatant contained v1.3-GFP protein, demonstrating the release of the intracellular receptor variant by this mechanism.
Collapse
Affiliation(s)
- Rauf Latif
- Thyroid Research Unit, Icahn School of Medicine at Mount Sinai, New York, New York
- James J. Peters VA Medical Center, New York, New York
- Correspondence: Rauf Latif, PhD, Icahn School of Medicine at Mount Sinai, Atran Berg 4-43, 1428 Madison Avenue, New York, New York 10029. E-mail:
| | - Mihaly Mezei
- Department of Structural and Chemical Biology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Syed A Morshed
- Thyroid Research Unit, Icahn School of Medicine at Mount Sinai, New York, New York
- James J. Peters VA Medical Center, New York, New York
| | - Risheng Ma
- Thyroid Research Unit, Icahn School of Medicine at Mount Sinai, New York, New York
- James J. Peters VA Medical Center, New York, New York
| | - Rachel Ehrlich
- Thyroid Research Unit, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Terry F Davies
- Thyroid Research Unit, Icahn School of Medicine at Mount Sinai, New York, New York
- James J. Peters VA Medical Center, New York, New York
| |
Collapse
|
20
|
Phylogenetic, molecular evolution and structural analyses of the WFDC1/prostate stromal protein 20 (ps20). Gene 2019; 686:125-140. [DOI: 10.1016/j.gene.2018.10.046] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 09/07/2018] [Accepted: 10/19/2018] [Indexed: 12/20/2022]
|
21
|
Singh P, Singh I, Shah K. Reduced Activity of Nitrate Reductase Under Heavy Metal Cadmium Stress in Rice: An in silico Answer. FRONTIERS IN PLANT SCIENCE 2019; 9:1948. [PMID: 30697220 PMCID: PMC6341063 DOI: 10.3389/fpls.2018.01948] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 12/14/2018] [Indexed: 05/26/2023]
Abstract
Cadmium is a well known toxic heavy metal, which has various detrimental effects on plant system. In plants an important enzyme involved in the production of nitric oxide, nitrate reductase, is also affected by cadmium toxicity. According to many studies cadmium has an inhibitory effect on nitrate reductase activity. Similar effect of cadmium was found in our study where an inhibitory effect of cadmium on nitrate reductase activity was noted. However, the mechanism behind this inhibition has not been explored. With the help of homology, 3-D structure of rice-nitrate reductase is modeled in this study. Its binding with nitrate, nitrite and cadmium metal in silico has been explored. The bonds formed between the enzyme-substrate complex, enzyme-cadmium and differences in interactions in presence of cadmium has been studied in detail. The present study should help in understanding the modeled structure of rice-nitrate reductase in 3-D which may in turn guide enzyme related studies in silico. The present study also provides an insight as to how cadmium interacts with nitrate reductase to alter the enzyme activity.
Collapse
Affiliation(s)
- Prerna Singh
- Department of Biochemistry, Faculty of Science, Banaras Hindu University, Varanasi, India
| | - Indra Singh
- Faculty of Science, School of Biotechnology, Banaras Hindu University, Varanasi, India
| | - Kavita Shah
- Institute of Environment and Sustainable Development, Banaras Hindu University, Varanasi, India
| |
Collapse
|
22
|
Pisupati A, Mickolajczyk KJ, Horton W, van Rossum DB, Anishkin A, Chintapalli SV, Li X, Chu-Luo J, Busey G, Hancock WO, Jegla T. The S6 gate in regulatory Kv6 subunits restricts heteromeric K + channel stoichiometry. J Gen Physiol 2018; 150:1702-1721. [PMID: 30322883 PMCID: PMC6279357 DOI: 10.1085/jgp.201812121] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 07/03/2018] [Accepted: 09/26/2018] [Indexed: 11/24/2022] Open
Abstract
Atypical substitutions in the S6 activation gate sequence distinguish “regulatory” Kv subunits, which cannot homotetramerize due to T1 self-incompatibility. Pisupati et al. show that such substitutions in Kv6 work together with self-incompatibility to restrict Kv2:Kv6 heteromeric stoichiometry to 3:1. The Shaker-like family of voltage-gated K+ channels comprises four functionally independent gene subfamilies, Shaker (Kv1), Shab (Kv2), Shaw (Kv3), and Shal (Kv4), each of which regulates distinct aspects of neuronal excitability. Subfamily-specific assembly of tetrameric channels is mediated by the N-terminal T1 domain and segregates Kv1–4, allowing multiple channel types to function independently in the same cell. Typical Shaker-like Kv subunits can form functional channels as homotetramers, but a group of mammalian Kv2-related genes (Kv5.1, Kv6s, Kv8s, and Kv9s) encodes subunits that have a “silent” or “regulatory” phenotype characterized by T1 self-incompatibility. These channels are unable to form homotetramers, but instead heteromerize with Kv2.1 or Kv2.2 to diversify the functional properties of these delayed rectifiers. While T1 self-incompatibility predicts that these heterotetramers could contain up to two regulatory (R) subunits, experiments show a predominance of 3:1R stoichiometry in which heteromeric channels contain a single regulatory subunit. Substitution of the self-compatible Kv2.1 T1 domain into the regulatory subunit Kv6.4 does not alter the stoichiometry of Kv2.1:Kv6.4 heteromers. Here, to identify other channel structures that might be responsible for favoring the 3:1R stoichiometry, we compare the sequences of mammalian regulatory subunits to independently evolved regulatory subunits from cnidarians. The most widespread feature of regulatory subunits is the presence of atypical substitutions in the highly conserved consensus sequence of the intracellular S6 activation gate of the pore. We show that two amino acid substitutions in the S6 gate of the regulatory subunit Kv6.4 restrict the functional stoichiometry of Kv2.1:Kv6.4 to 3:1R by limiting the formation and function of 2:2R heteromers. We propose a two-step model for the evolution of the asymmetric 3:1R stoichiometry, which begins with evolution of self-incompatibility to establish the regulatory phenotype, followed by drift of the activation gate consensus sequence under relaxed selection to limit stoichiometry to 3:1R.
Collapse
Affiliation(s)
- Aditya Pisupati
- Department of Biology, Pennsylvania State University, University Park, PA.,Medical Scientist Training Program, College of Medicine, Pennsylvania State University, Hershey, PA
| | - Keith J Mickolajczyk
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA
| | - William Horton
- Department of Animal Science, Pennsylvania State University, University Park, PA
| | - Damian B van Rossum
- The Jake Gittlen Laboratories for Cancer Research, College of Medicine, Pennsylvania State University, Hershey, PA.,Division of Experimental Pathology, Department of Pathology, College of Medicine, Pennsylvania State University, Hershey, PA
| | - Andriy Anishkin
- Department of Biology, University of Maryland, College Park, MD
| | - Sree V Chintapalli
- Arkansas Children's Nutrition Center and Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR
| | - Xiaofan Li
- Department of Biology, Pennsylvania State University, University Park, PA
| | - Jose Chu-Luo
- Department of Biology, Pennsylvania State University, University Park, PA
| | - Gregory Busey
- Department of Biology, Pennsylvania State University, University Park, PA
| | - William O Hancock
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA
| | - Timothy Jegla
- Department of Biology, Pennsylvania State University, University Park, PA .,Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA
| |
Collapse
|
23
|
Sagratini G, Buccioni M, Marucci G, Poggesi E, Skorski M, Costanzi S, Giardinà D. Chiral analogues of (+)-cyclazosin as potent α 1B-adrenoceptor selective antagonist. Bioorg Med Chem 2018; 26:3502-3513. [PMID: 29784274 DOI: 10.1016/j.bmc.2018.05.023] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 05/11/2018] [Accepted: 05/16/2018] [Indexed: 12/21/2022]
Abstract
(+)-Cyclazosin [(+)-1] is one of most selective antagonists of the α1B-adrenoceptor subtype (selectivity ratios, α1B/α1A = 13, α1B/α1D = 38-39). To improve the selectivity, we synthesized and pharmacologically studied the blocking activity against α1-adrenoceptors of several homochiral analogues of (+)-cyclazosin featuring different substituents on the carbonyl or amine groups, namely (-)-2, (+)-3, (-)-4-(-)-8, (+)-9. Moreover, we studied the activity of some their opposite enantiomers, namely (-)-1, (-)-3, (+)-6, and (-)-9, to evaluate the influence of stereochemistry on selectivity. The benzyloxycarbonyl and methyl (4aS,8aR) analogues (+)-3 and (-)-6 improved in a significant way the α1B selectivity of the progenitor compound: 4 and 14 time vs. the α1D subtype and 35 and 77 times vs. the α1A subtype, respectively. The study confirmed the importance of the hydrophobic cis-octahydroquinoxaline moiety of these molecules for the establishment of interactions with the α1-adrenoceptors as well that of their (4aS,8aR) stereochemistry to grant selectivity for the α1B subtype. Hypotheses on the mode of interaction of these compounds were advanced on the basis of molecular modeling studies performed on compound (+)-3.
Collapse
Affiliation(s)
- Gianni Sagratini
- Scuola in Scienze del Farmaco e dei Prodotti della Salute, Università di Camerino, Via S. Agostino 1, 62032 Camerino, Italy.
| | - Michela Buccioni
- Scuola in Scienze del Farmaco e dei Prodotti della Salute, Università di Camerino, Via S. Agostino 1, 62032 Camerino, Italy
| | - Gabriella Marucci
- Scuola in Scienze del Farmaco e dei Prodotti della Salute, Università di Camerino, Via S. Agostino 1, 62032 Camerino, Italy
| | - Elena Poggesi
- Drug Discovery Division, Recordati SpA, Via Civitali 1, 20148 Milano, Italy
| | - Matthew Skorski
- Department of Chemistry, American University, Washington, DC 20016, USA
| | - Stefano Costanzi
- Department of Chemistry, American University, Washington, DC 20016, USA; Center for Behavioral Neuroscience, American University, Washington, DC 20016, USA
| | - Dario Giardinà
- Scuola in Scienze del Farmaco e dei Prodotti della Salute, Università di Camerino, Via S. Agostino 1, 62032 Camerino, Italy
| |
Collapse
|
24
|
Broadly conserved Na +-binding site in the N-lobe of prokaryotic multidrug MATE transporters. Proc Natl Acad Sci U S A 2018; 115:E6172-E6181. [PMID: 29915058 DOI: 10.1073/pnas.1802080115] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Multidrug and toxic-compound extrusion (MATE) proteins comprise an important but largely uncharacterized family of secondary-active transporters. In both eukaryotes and prokaryotes, these transporters protect the cell by catalyzing the efflux of a broad range of cytotoxic compounds, including human-made antibiotics and anticancer drugs. MATEs are thus potential pharmacological targets against drug-resistant pathogenic bacteria and tumor cells. The activity of MATEs is powered by transmembrane electrochemical ion gradients, but their molecular mechanism and ion specificity are not understood, in part because high-quality structural information is limited. Here, we use computational methods to study PfMATE, from Pyrococcus furiosus, whose structure is the best resolved to date. Analysis of available crystallographic data and additional molecular dynamics simulations unequivocally reveal an occupied Na+-binding site in the N-lobe of this transporter, which had not been previously recognized. We find this site to be selective against K+ and broadly conserved among prokaryotic MATEs, including homologs known to be Na+-dependent such as NorM-VC, VmrA, and ClbM, for which the location of the Na+ site had been debated. We note, however, that the chemical makeup of the proposed Na+ site indicates it is weakly specific against H+, explaining why MATEs featuring this Na+-binding motif may be solely driven by H+ in laboratory conditions. We further posit that the concurrent coupling to H+ and Na+ gradients observed for some Na+-driven MATEs owes to a second H+-binding site, within the C-lobe. In summary, our study provides insights into the structural basis for the complex ion dependency of MATE transporters.
Collapse
|
25
|
Cacciotto P, Ramaswamy VK, Malloci G, Ruggerone P, Vargiu AV. Molecular Modeling of Multidrug Properties of Resistance Nodulation Division (RND) Transporters. Methods Mol Biol 2018; 1700:179-219. [PMID: 29177832 DOI: 10.1007/978-1-4939-7454-2_11] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Efflux pumps of the resistance nodulation division (RND) superfamily are among the major contributors to intrinsic and acquired multidrug resistance in Gram-negative bacteria. Structural information on AcrAB-TolC and MexAB-OprM, major efflux pumps of Escherichia coli and Pseudomonas aeruginosa respectively, boosted intensive research aimed at understanding the molecular mechanisms ruling the active extrusion processes. In particular, several studies were devoted to the understanding of the determinants behind the extraordinary broad specificity of the RND transporters AcrB and MexB. In this chapter, we discuss the ever-growing role computational methods have been playing in deciphering key structural and dynamical features of these transporters and of their interaction with substrates and inhibitors. We further discuss and illustrate examples from our lab of how molecular docking, homology modeling, all-atom molecular dynamics simulations and in silico free energy estimations can all together give precious insights into the processes of recognition and extrusion of substrates, as well as on the possible inhibition strategies.
Collapse
Affiliation(s)
- Pierpaolo Cacciotto
- Department of Physics, University of Cagliari, s.p. 8, 09042, Monserrato, CA, Italy
| | - Venkata K Ramaswamy
- Department of Physics, University of Cagliari, s.p. 8, 09042, Monserrato, CA, Italy
| | - Giuliano Malloci
- Department of Physics, University of Cagliari, s.p. 8, 09042, Monserrato, CA, Italy
| | - Paolo Ruggerone
- Department of Physics, University of Cagliari, s.p. 8, 09042, Monserrato, CA, Italy
| | - Attilio V Vargiu
- Department of Physics, University of Cagliari, s.p. 8, 09042, Monserrato, CA, Italy.
| |
Collapse
|
26
|
Kaiser TM, Kell SA, Kusumoto H, Shaulsky G, Bhattacharya S, Epplin MP, Strong KL, Miller EJ, Cox BD, Menaldino DS, Liotta DC, Traynelis SF, Burger PB. The Bioactive Protein-Ligand Conformation of GluN2C-Selective Positive Allosteric Modulators Bound to the NMDA Receptor. Mol Pharmacol 2017; 93:141-156. [PMID: 29242355 DOI: 10.1124/mol.117.110940] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 12/11/2017] [Indexed: 12/18/2022] Open
Abstract
N-methyl-d-aspartate (NMDA) receptors are ligand-gated, cation-selective channels that mediate a slow component of excitatory synaptic transmission. Subunit-selective positive allosteric modulators of NMDA receptor function have therapeutically relevant effects on multiple processes in the brain. A series of pyrrolidinones, such as PYD-106, that selectively potentiate NMDA receptors that contain the GluN2C subunit have structural determinants of activity that reside between the GluN2C amino terminal domain and the GluN2C agonist binding domain, suggesting a unique site of action. Here we use molecular biology and homology modeling to identify residues that line a candidate binding pocket for GluN2C-selective pyrrolidinones. We also show that occupancy of only one site in diheteromeric receptors is required for potentiation. Both GluN2A and GluN2B can dominate the sensitivity of triheteromeric receptors to eliminate the actions of pyrrolidinones, thus rendering this series uniquely sensitive to subunit stoichiometry. We experimentally identified NMR-derived conformers in solution, which combined with molecular modeling allows the prediction of the bioactive binding pose for this series of GluN2C-selective positive allosteric modulators of NMDA receptors. These data advance our understanding of the site and nature of the ligand-protein interaction for GluN2C-selective positive allosteric modulators for NMDA receptors.
Collapse
Affiliation(s)
- Thomas M Kaiser
- Department of Chemistry, Emory University, Atlanta, Georgia (T.M.K., S.A.K., M.P.E., K.L.S., E.J.M., B.D.C., D.S.M., D.C.L., P.B.B.); and Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia (H.K., G.S., S.B., S.F.T.)
| | - Steven A Kell
- Department of Chemistry, Emory University, Atlanta, Georgia (T.M.K., S.A.K., M.P.E., K.L.S., E.J.M., B.D.C., D.S.M., D.C.L., P.B.B.); and Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia (H.K., G.S., S.B., S.F.T.)
| | - Hirofumi Kusumoto
- Department of Chemistry, Emory University, Atlanta, Georgia (T.M.K., S.A.K., M.P.E., K.L.S., E.J.M., B.D.C., D.S.M., D.C.L., P.B.B.); and Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia (H.K., G.S., S.B., S.F.T.)
| | - Gil Shaulsky
- Department of Chemistry, Emory University, Atlanta, Georgia (T.M.K., S.A.K., M.P.E., K.L.S., E.J.M., B.D.C., D.S.M., D.C.L., P.B.B.); and Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia (H.K., G.S., S.B., S.F.T.)
| | - Subhrajit Bhattacharya
- Department of Chemistry, Emory University, Atlanta, Georgia (T.M.K., S.A.K., M.P.E., K.L.S., E.J.M., B.D.C., D.S.M., D.C.L., P.B.B.); and Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia (H.K., G.S., S.B., S.F.T.)
| | - Matthew P Epplin
- Department of Chemistry, Emory University, Atlanta, Georgia (T.M.K., S.A.K., M.P.E., K.L.S., E.J.M., B.D.C., D.S.M., D.C.L., P.B.B.); and Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia (H.K., G.S., S.B., S.F.T.)
| | - Katie L Strong
- Department of Chemistry, Emory University, Atlanta, Georgia (T.M.K., S.A.K., M.P.E., K.L.S., E.J.M., B.D.C., D.S.M., D.C.L., P.B.B.); and Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia (H.K., G.S., S.B., S.F.T.)
| | - Eric J Miller
- Department of Chemistry, Emory University, Atlanta, Georgia (T.M.K., S.A.K., M.P.E., K.L.S., E.J.M., B.D.C., D.S.M., D.C.L., P.B.B.); and Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia (H.K., G.S., S.B., S.F.T.)
| | - Bryan D Cox
- Department of Chemistry, Emory University, Atlanta, Georgia (T.M.K., S.A.K., M.P.E., K.L.S., E.J.M., B.D.C., D.S.M., D.C.L., P.B.B.); and Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia (H.K., G.S., S.B., S.F.T.)
| | - David S Menaldino
- Department of Chemistry, Emory University, Atlanta, Georgia (T.M.K., S.A.K., M.P.E., K.L.S., E.J.M., B.D.C., D.S.M., D.C.L., P.B.B.); and Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia (H.K., G.S., S.B., S.F.T.)
| | - Dennis C Liotta
- Department of Chemistry, Emory University, Atlanta, Georgia (T.M.K., S.A.K., M.P.E., K.L.S., E.J.M., B.D.C., D.S.M., D.C.L., P.B.B.); and Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia (H.K., G.S., S.B., S.F.T.)
| | - Stephen F Traynelis
- Department of Chemistry, Emory University, Atlanta, Georgia (T.M.K., S.A.K., M.P.E., K.L.S., E.J.M., B.D.C., D.S.M., D.C.L., P.B.B.); and Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia (H.K., G.S., S.B., S.F.T.)
| | - Pieter B Burger
- Department of Chemistry, Emory University, Atlanta, Georgia (T.M.K., S.A.K., M.P.E., K.L.S., E.J.M., B.D.C., D.S.M., D.C.L., P.B.B.); and Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia (H.K., G.S., S.B., S.F.T.)
| |
Collapse
|
27
|
Ficici E, Faraldo-Gómez JD, Jennings ML, Forrest LR. Asymmetry of inverted-topology repeats in the AE1 anion exchanger suggests an elevator-like mechanism. J Gen Physiol 2017; 149:1149-1164. [PMID: 29167180 PMCID: PMC5715908 DOI: 10.1085/jgp.201711836] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 10/10/2017] [Indexed: 12/02/2022] Open
Abstract
Anion exchanger 1 catalyzes the transmembrane antiport of chloride and bicarbonate ions through a mechanism that has remained unclear. By modeling its inward-facing state and comparing it with the known outward-facing form, Ficici et al. hypothesize that this transporter features an elevator-like mechanism. The membrane transporter anion exchanger 1 (AE1), or band 3, is a key component in the processes of carbon-dioxide transport in the blood and urinary acidification in the renal collecting duct. In both erythrocytes and the basolateral membrane of the collecting-duct α-intercalated cells, the role of AE1 is to catalyze a one-for-one exchange of chloride for bicarbonate. After decades of biochemical and functional studies, the structure of the transmembrane region of AE1, which catalyzes the anion-exchange reaction, has finally been determined. Each protomer of the AE1 dimer comprises two repeats with inverted transmembrane topologies, but the structures of these repeats differ. This asymmetry causes the putative substrate-binding site to be exposed only to the extracellular space, consistent with the expectation that anion exchange occurs via an alternating-access mechanism. Here, we hypothesize that the unknown, inward-facing conformation results from inversion of this asymmetry, and we propose a model of this state constructed using repeat-swap homology modeling. By comparing this inward-facing model with the outward-facing experimental structure, we predict that the mechanism of AE1 involves an elevator-like motion of the substrate-binding domain relative to the nearly stationary dimerization domain and to the membrane plane. This hypothesis is in qualitative agreement with a wide range of biochemical and functional data, which we review in detail, and suggests new avenues of experimentation.
Collapse
Affiliation(s)
- Emel Ficici
- Theoretical Molecular Biophysics Laboratory, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| | - José D Faraldo-Gómez
- Theoretical Molecular Biophysics Laboratory, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Michael L Jennings
- Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, Little Rock, AR
| | - Lucy R Forrest
- Computational Structural Biology Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD
| |
Collapse
|
28
|
Solís-Calero C, Carvalho HF. KLK14 interactions with HAI-1 and HAI-2 serine protease inhibitors: A molecular dynamics and relative free-energy calculations study. Cell Biol Int 2017; 41:1246-1264. [PMID: 28817220 DOI: 10.1002/cbin.10839] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 08/12/2017] [Indexed: 01/13/2023]
Abstract
Kallikrein 14 (KLK14) is a serine protease linked to several pathologies including prostate cancer and positively correlates with Gleason score. Though KLK14 functioning in cancer is poorly understood, it has been implicated in HGF/Met signaling, given that KLK14 proteolytically inhibits HGF activator-inhibitor 1 (HAI-1), which strongly inhibits pro-HGF activators, thereby contributing to tumor progression. In this work, KLK14 binding to either hepatocyte growth factor activator inhibitor type-1 (HAI-1) or type-2 (HAI-2) was essayed using homology modeling, molecular dynamic simulations and free-energy calculations through MM/PBSA and MM/GBSA. KLK14 was successfully modeled. Calculated free energies suggested higher binding affinity for the KLK14/HAI-1 interaction than for KLK14/HAI-2. This difference in binding affinity is largely explained by the higher stability of the hydrogen-bond networks in KLK14/HAI-1 along the simulation trajectory. A key arginine residue in both HAI-1 and HAI-2 is responsible for their interaction with the S1 pocket in KLK14. Additionally, MM/GBSA free-energy decomposition postulates that KLK14 Asp174 and Trp196 are hotspots for binding HAI-1 and HAI-2.
Collapse
Affiliation(s)
- Christian Solís-Calero
- Department of Structural and Functional Biology, State University of Campinas, Campinas, São Paulo, Brazil
| | - Hernandes F Carvalho
- Department of Structural and Functional Biology, State University of Campinas, Campinas, São Paulo, Brazil
| |
Collapse
|
29
|
Kacar B, Hanson‐Smith V, Adam ZR, Boekelheide N. Constraining the timing of the Great Oxidation Event within the Rubisco phylogenetic tree. GEOBIOLOGY 2017; 15:628-640. [PMID: 28670785 PMCID: PMC5575542 DOI: 10.1111/gbi.12243] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 05/09/2017] [Indexed: 05/04/2023]
Abstract
Ribulose 1,5-bisphosphate (RuBP) carboxylase/oxygenase (RuBisCO, or Rubisco) catalyzes a key reaction by which inorganic carbon is converted into organic carbon in the metabolism of many aerobic and anaerobic organisms. Across the broader Rubisco protein family, homologs exhibit diverse biochemical characteristics and metabolic functions, but the evolutionary origins of this diversity are unclear. Evidence of the timing of Rubisco family emergence and diversification of its different forms has been obscured by a meager paleontological record of early Earth biota, their subcellular physiology and metabolic components. Here, we use computational models to reconstruct a Rubisco family phylogenetic tree, ancestral amino acid sequences at branching points on the tree, and protein structures for several key ancestors. Analysis of historic substitutions with respect to their structural locations shows that there were distinct periods of amino acid substitution enrichment above background levels near and within its oxygen-sensitive active site and subunit interfaces over the divergence between Form III (associated with anoxia) and Form I (associated with oxia) groups in its evolutionary history. One possible interpretation is that these periods of substitutional enrichment are coincident with oxidative stress exerted by the rise of oxygenic photosynthesis in the Precambrian era. Our interpretation implies that the periods of Rubisco substitutional enrichment inferred near the transition from anaerobic Form III to aerobic Form I ancestral sequences predate the acquisition of Rubisco by fully derived cyanobacterial (i.e., dual photosystem-bearing, oxygen-evolving) clades. The partitioning of extant lineages at high clade levels within our Rubisco phylogeny indicates that horizontal transfer of Rubisco is a relatively infrequent event. Therefore, it is possible that the mutational enrichment periods between the Form III and Form I common ancestral sequences correspond to the adaptation of key oxygen-sensitive components of Rubisco prior to, or coincident with, the Great Oxidation Event.
Collapse
Affiliation(s)
- B. Kacar
- Department of Organismic and Evolutionary BiologyHarvard UniversityCambridgeMAUSA
| | - V. Hanson‐Smith
- Department of Microbiology and ImmunologyUniversity of California San FranciscoSan FranciscoCAUSA
| | - Z. R. Adam
- Department of Earth and Planetary SciencesHarvard UniversityCambridgeMAUSA
| | | |
Collapse
|
30
|
Kumar R, Maurya R, Saran S. Identification of novel inhibitors of the translationally controlled tumor protein (TCTP): insights from molecular dynamics. MOLECULAR BIOSYSTEMS 2017; 13:510-524. [PMID: 28128835 DOI: 10.1039/c6mb00850j] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The translationally controlled tumor protein (TCTP) is a highly conserved multifunctional protein, preferentially expressed in mitotically active tissues and is a potential biomarker and a therapeutic target for lung cancers. An understanding of the biology of this molecule and model systems for the screening of drugs is still awaited. In the absence of complete crystal structure, NMR structures as templates were used for homology modeling and MD optimization of both Dictyostelium discoideum and human TCTPs, which was followed by pocket-site prediction, ligand screening and docking. Rescoring of TCTP-ligand complexes was done using MD and MM-PBSA approaches. D. discoideum TCTP was expressed under a constitutive promoter and the endogenous RNA in multicellular structures formed was localized by in situ hybridization. Based on the interactions and binding energy scores, two novel compounds were identified as the best potential inhibitors that could be further used for the development of drug candidates. Inhibition of cell proliferation was observed in the strain overexpressing Dictyostelium TCTP and in situ hybridization results show them to be localized in the prestalk (dying cell population) cells. D. discoideum and human TCTPs share similar dynamic behaviors; overexpression of Dictyostelium TCTP inhibits cell proliferation. D. discoideum could be used as a model system for understanding the biology of this molecule and also for drug screening.
Collapse
Affiliation(s)
- Rakesh Kumar
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India.
| | - Ranjana Maurya
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India.
| | - Shweta Saran
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India.
| |
Collapse
|
31
|
Shimizu H, Tosaki A, Ohsawa N, Ishizuka-Katsura Y, Shoji S, Miyazaki H, Oyama F, Terada T, Shirouzu M, Sekine SI, Nukina N, Yokoyama S. Parallel homodimer structures of the extracellular domains of the voltage-gated sodium channel β4 subunit explain its role in cell-cell adhesion. J Biol Chem 2017; 292:13428-13440. [PMID: 28655765 PMCID: PMC5555201 DOI: 10.1074/jbc.m117.786509] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 06/26/2017] [Indexed: 11/06/2022] Open
Abstract
Voltage-gated sodium channels (VGSCs) are transmembrane proteins required for the generation of action potentials in excitable cells and essential for propagating electrical impulses along nerve cells. VGSCs are complexes of a pore-forming α subunit and auxiliary β subunits, designated as β1/β1B-β4 (encoded by SCN1B-4B, respectively), which also function in cell-cell adhesion. We previously reported the structural basis for the trans homophilic interaction of the β4 subunit, which contributes to its adhesive function. Here, using crystallographic and biochemical analyses, we show that the β4 extracellular domains directly interact with each other in a parallel manner that involves an intermolecular disulfide bond between the unpaired Cys residues (Cys58) in the loop connecting strands B and C and intermolecular hydrophobic and hydrogen-bonding interactions of the N-terminal segments (Ser30-Val35). Under reducing conditions, an N-terminally deleted β4 mutant exhibited decreased cell adhesion compared with the wild type, indicating that the β4 cis dimer contributes to the trans homophilic interaction of β4 in cell-cell adhesion. Furthermore, this mutant exhibited increased association with the α subunit, indicating that the cis dimerization of β4 affects α-β4 complex formation. These observations provide the structural basis for the parallel dimer formation of β4 in VGSCs and reveal its mechanism in cell-cell adhesion.
Collapse
Affiliation(s)
- Hideaki Shimizu
- From the RIKEN Systems and Structural Biology Center, Tsurumi, Yokohama 230-0045, Japan.,the RIKEN Center for Life Science Technologies, Tsurumi, Yokohama 230-0045, Japan.,the Laboratory for Structural Neuropathology, RIKEN Brain Science Institute, Wako, Saitama 351-0198, Japan
| | - Asako Tosaki
- the Laboratory for Structural Neuropathology, RIKEN Brain Science Institute, Wako, Saitama 351-0198, Japan
| | - Noboru Ohsawa
- From the RIKEN Systems and Structural Biology Center, Tsurumi, Yokohama 230-0045, Japan.,the RIKEN Center for Life Science Technologies, Tsurumi, Yokohama 230-0045, Japan
| | - Yoshiko Ishizuka-Katsura
- From the RIKEN Systems and Structural Biology Center, Tsurumi, Yokohama 230-0045, Japan.,the RIKEN Center for Life Science Technologies, Tsurumi, Yokohama 230-0045, Japan
| | - Shisako Shoji
- From the RIKEN Systems and Structural Biology Center, Tsurumi, Yokohama 230-0045, Japan.,the RIKEN Center for Life Science Technologies, Tsurumi, Yokohama 230-0045, Japan
| | - Haruko Miyazaki
- the Laboratory for Structural Neuropathology, RIKEN Brain Science Institute, Wako, Saitama 351-0198, Japan.,the Department of Neuroscience for Neurodegenerative Disorders, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan.,the Laboratory of Structural Neuropathology, Doshisha University Graduate School of Brain Science, 1-3 Tatara Miyakodani, Kyotanabe-shi, Kyoto 610-0394, Japan
| | - Fumitaka Oyama
- the Laboratory for Structural Neuropathology, RIKEN Brain Science Institute, Wako, Saitama 351-0198, Japan.,the Department of Chemistry and Life Science, Kogakuin University, Hachioji, Tokyo 192-0015, Japan, and
| | - Takaho Terada
- From the RIKEN Systems and Structural Biology Center, Tsurumi, Yokohama 230-0045, Japan.,the RIKEN Structural Biology Laboratory, Tsurumi, Yokohama 230-0045, Japan
| | - Mikako Shirouzu
- From the RIKEN Systems and Structural Biology Center, Tsurumi, Yokohama 230-0045, Japan.,the RIKEN Center for Life Science Technologies, Tsurumi, Yokohama 230-0045, Japan
| | - Shun-Ichi Sekine
- From the RIKEN Systems and Structural Biology Center, Tsurumi, Yokohama 230-0045, Japan.,the RIKEN Center for Life Science Technologies, Tsurumi, Yokohama 230-0045, Japan
| | - Nobuyuki Nukina
- the Laboratory for Structural Neuropathology, RIKEN Brain Science Institute, Wako, Saitama 351-0198, Japan.,the Department of Neuroscience for Neurodegenerative Disorders, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan.,the Laboratory of Structural Neuropathology, Doshisha University Graduate School of Brain Science, 1-3 Tatara Miyakodani, Kyotanabe-shi, Kyoto 610-0394, Japan
| | - Shigeyuki Yokoyama
- From the RIKEN Systems and Structural Biology Center, Tsurumi, Yokohama 230-0045, Japan, .,the RIKEN Structural Biology Laboratory, Tsurumi, Yokohama 230-0045, Japan
| |
Collapse
|
32
|
Cloning, expression, purification and biophysical analysis of two putative halogenases from the glycopeptide A47,934 gene cluster of Streptomyces toyocaensis. Protein Expr Purif 2017; 132:9-18. [DOI: 10.1016/j.pep.2017.01.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 12/22/2016] [Accepted: 01/04/2017] [Indexed: 10/20/2022]
|
33
|
Chen W, Tankovic A, Burger PB, Kusumoto H, Traynelis SF, Yuan H. Functional Evaluation of a De Novo GRIN2A Mutation Identified in a Patient with Profound Global Developmental Delay and Refractory Epilepsy. Mol Pharmacol 2017; 91:317-330. [PMID: 28126851 DOI: 10.1124/mol.116.106781] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2016] [Accepted: 01/23/2017] [Indexed: 12/22/2022] Open
Abstract
The N-methyl-d-aspartate receptor (NMDAR), a ligand-gated ionotropic glutamate receptor, plays important roles in normal brain development and a wide range of neurologic disorders, including epilepsy. Here, we evaluate for the first time the functional properties of a de novo GRIN2A missense mutation (p.M817V) in the pre-M4 linker in a child with profound global developmental delay and refractory epilepsy. Electrophysiologic recordings revealed that the mutant GluN2A(M817V)-containing receptors showed enhanced agonist potency, reduced sensitivity to endogenous negative inhibitors (Mg2+, proton, and zinc), prolonged synaptic-like response time course, increased single-channel mean open time, and increased channel open probability. These results suggest that the gain-of-function M817V mutation causes overactivation of NMDAR and drives neuronal hyperexcitability, which may contribute to the patient's observed epileptic phenotype. Molecular modeling of the closed channel conformation reveals that this mutation weakens the interaction between GluN2 transmembrane helix M4 and two GluN1 transmembrane helices, and increases atomic fluctuation or movement of the pre-M1 region of GluN1 subunit, suggesting a mechanism by which channel function is enhanced. The functional changes of this mutation on agonist potency occur when the mutation is introduced into all other GluN2 subunits, suggesting a conserved role of this residue in control of NMDAR function through interactions of membrane spanning GluN2 and GluN1 helices. A number of NMDAR-targeted drugs including U.S. Food and Drug Association-approved NMDAR channel blockers were evaluated for their ability to inhibit receptors containing GluN2A(M817V) as a first step to exploring the potential for rescue pharmacology and personalized medicine.
Collapse
Affiliation(s)
- Wenjuan Chen
- Department of Pharmacology, Rollins Research Center, School of Medicine, Emory University, Atlanta, Georgia (W.C., A.T., H.K., S.F.T., H.Y.); Department of Neurology, Xiangya Hospital, Central South University, Changsha, People's Republic of China (W.C.); Department of Chemistry, Emory University, Atlanta, Georgia (P.B.B.); Center for Functional Evaluation of Rare Variants (CFERV), Rollins Research Center, School of Medicine, Emory University, Atlanta, Georgia (S.F.T., H.Y.)
| | - Anel Tankovic
- Department of Pharmacology, Rollins Research Center, School of Medicine, Emory University, Atlanta, Georgia (W.C., A.T., H.K., S.F.T., H.Y.); Department of Neurology, Xiangya Hospital, Central South University, Changsha, People's Republic of China (W.C.); Department of Chemistry, Emory University, Atlanta, Georgia (P.B.B.); Center for Functional Evaluation of Rare Variants (CFERV), Rollins Research Center, School of Medicine, Emory University, Atlanta, Georgia (S.F.T., H.Y.)
| | - Pieter B Burger
- Department of Pharmacology, Rollins Research Center, School of Medicine, Emory University, Atlanta, Georgia (W.C., A.T., H.K., S.F.T., H.Y.); Department of Neurology, Xiangya Hospital, Central South University, Changsha, People's Republic of China (W.C.); Department of Chemistry, Emory University, Atlanta, Georgia (P.B.B.); Center for Functional Evaluation of Rare Variants (CFERV), Rollins Research Center, School of Medicine, Emory University, Atlanta, Georgia (S.F.T., H.Y.)
| | - Hirofumi Kusumoto
- Department of Pharmacology, Rollins Research Center, School of Medicine, Emory University, Atlanta, Georgia (W.C., A.T., H.K., S.F.T., H.Y.); Department of Neurology, Xiangya Hospital, Central South University, Changsha, People's Republic of China (W.C.); Department of Chemistry, Emory University, Atlanta, Georgia (P.B.B.); Center for Functional Evaluation of Rare Variants (CFERV), Rollins Research Center, School of Medicine, Emory University, Atlanta, Georgia (S.F.T., H.Y.)
| | - Stephen F Traynelis
- Department of Pharmacology, Rollins Research Center, School of Medicine, Emory University, Atlanta, Georgia (W.C., A.T., H.K., S.F.T., H.Y.); Department of Neurology, Xiangya Hospital, Central South University, Changsha, People's Republic of China (W.C.); Department of Chemistry, Emory University, Atlanta, Georgia (P.B.B.); Center for Functional Evaluation of Rare Variants (CFERV), Rollins Research Center, School of Medicine, Emory University, Atlanta, Georgia (S.F.T., H.Y.)
| | - Hongjie Yuan
- Department of Pharmacology, Rollins Research Center, School of Medicine, Emory University, Atlanta, Georgia (W.C., A.T., H.K., S.F.T., H.Y.); Department of Neurology, Xiangya Hospital, Central South University, Changsha, People's Republic of China (W.C.); Department of Chemistry, Emory University, Atlanta, Georgia (P.B.B.); Center for Functional Evaluation of Rare Variants (CFERV), Rollins Research Center, School of Medicine, Emory University, Atlanta, Georgia (S.F.T., H.Y.)
| |
Collapse
|
34
|
Weth A, Dippl C, Striedner Y, Tiemann-Boege I, Vereshchaga Y, Golenhofen N, Bartelt-Kirbach B, Baumgartner W. Water transport through the intestinal epithelial barrier under different osmotic conditions is dependent on LI-cadherin trans-interaction. Tissue Barriers 2017; 5:e1285390. [PMID: 28452574 PMCID: PMC5501135 DOI: 10.1080/21688370.2017.1285390] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
In the intestine water has to be reabsorbed from the chymus across the intestinal epithelium. The osmolarity within the lumen is subjected to high variations meaning that water transport often has to take place against osmotic gradients. It has been hypothesized that LI-cadherin is important in this process by keeping the intercellular cleft narrow facilitating the buildup of an osmotic gradient allowing water reabsorption. LI-cadherin is exceptional among the cadherin superfamily with respect to its localization along the lateral plasma membrane of epithelial cells being excluded from adherens junction. Furthermore it has 7 but not 5 extracellular cadherin repeats (EC1-EC7) and a small cytosolic domain. In this study we identified the peptide VAALD as an inhibitor of LI-cadherin trans-interaction by modeling the structure of LI-cadherin and comparison with the known adhesive interfaces of E-cadherin. This inhibitory peptide was used to measure LI-cadherin dependency of water transport through a monolayer of epithelial CACO2 cells under various osmotic conditions. If LI-cadherin trans-interaction was inhibited by use of the peptide, water transport from the luminal to the basolateral side was impaired and even reversed in the case of hypertonic conditions whereas no effect could be observed at isotonic conditions. These data are in line with a recently published model predicting LI-cadherin to keep the width of the lateral intercellular cleft small. In this narrow cleft a high osmolarity can be achieved due to ion pumps yielding a standing osmotic gradient allowing water absorption from the gut even if the faeces is highly hypertonic.
Collapse
Affiliation(s)
- Agnes Weth
- a Institute of Biomedical Mechatronics, Johannes Kepler University of Linz , Linz , Austria
| | - Carsten Dippl
- a Institute of Biomedical Mechatronics, Johannes Kepler University of Linz , Linz , Austria
| | - Yasmin Striedner
- b Institute of Biophysics, Johannes Kepler University of Linz , Linz , Austria
| | - Irene Tiemann-Boege
- b Institute of Biophysics, Johannes Kepler University of Linz , Linz , Austria
| | - Yana Vereshchaga
- a Institute of Biomedical Mechatronics, Johannes Kepler University of Linz , Linz , Austria
| | - Nikola Golenhofen
- c Institute of Anatomy and Cell Biology, University of Ulm , Ulm , Germany
| | | | - Werner Baumgartner
- a Institute of Biomedical Mechatronics, Johannes Kepler University of Linz , Linz , Austria
| |
Collapse
|
35
|
Abstract
Genome sequencing projects have resulted in a rapid increase in the number of known protein sequences. In contrast, only about one-hundredth of these sequences have been characterized at atomic resolution using experimental structure determination methods. Computational protein structure modeling techniques have the potential to bridge this sequence-structure gap. In the following chapter, we present an example that illustrates the use of MODELLER to construct a comparative model for a protein with unknown structure. Automation of a similar protocol has resulted in models of useful accuracy for domains in more than half of all known protein sequences.
Collapse
Affiliation(s)
- Benjamin Webb
- Department of Bioengineering and Therapeutic Sciences, Department of Pharmaceutical Chemistry, and California Institute for Quantitative Biosciences (QB3), University of California San Francisco, San Francisco, CA, 94143, USA
| | - Andrej Sali
- Department of Bioengineering and Therapeutic Sciences, Department of Pharmaceutical Chemistry, and California Institute for Quantitative Biosciences (QB3), University of California San Francisco, San Francisco, CA, 94143, USA.
| |
Collapse
|
36
|
Sima M, Novotny M, Pravda L, Sumova P, Rohousova I, Volf P. The Diversity of Yellow-Related Proteins in Sand Flies (Diptera: Psychodidae). PLoS One 2016; 11:e0166191. [PMID: 27812196 PMCID: PMC5094789 DOI: 10.1371/journal.pone.0166191] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 10/24/2016] [Indexed: 01/06/2023] Open
Abstract
Yellow-related proteins (YRPs) present in sand fly saliva act as affinity binders of bioamines, and help the fly to complete a bloodmeal by scavenging the physiological signals of damaged cells. They are also the main antigens in sand fly saliva and their recombinant form is used as a marker of host exposure to sand flies. Moreover, several salivary proteins and plasmids coding these proteins induce strong immune response in hosts bitten by sand flies and are being used to design protecting vaccines against Leishmania parasites. In this study, thirty two 3D models of different yellow-related proteins from thirteen sand fly species of two genera were constructed based on the known protein structure from Lutzomyia longipalpis. We also studied evolutionary relationships among species based on protein sequences as well as sequence and structural variability of their ligand-binding site. All of these 33 sand fly YRPs shared a similar structure, including a unique tunnel that connects the ligand-binding site with the solvent by two independent paths. However, intraspecific modifications found among these proteins affects the charges of the entrances to the tunnel, the length of the tunnel and its hydrophobicity. We suggest that these structural and sequential differences influence the ligand-binding abilities of these proteins and provide sand flies with a greater number of YRP paralogs with more nuanced answers to bioamines. All these characteristics allow us to better evaluate these proteins with respect to their potential use as part of anti-Leishmania vaccines or as an antigen to measure host exposure to sand flies.
Collapse
Affiliation(s)
- Michal Sima
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czech Republic
- * E-mail:
| | - Marian Novotny
- Department of Cell Biology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Lukas Pravda
- CEITEC—Central European Institute of Technology, Masaryk University, Brno, Czech Republic
- National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Petra Sumova
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Iva Rohousova
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Petr Volf
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czech Republic
| |
Collapse
|
37
|
Abstract
Comparative protein structure modeling predicts the three-dimensional structure of a given protein sequence (target) based primarily on its alignment to one or more proteins of known structure (templates). The prediction process consists of fold assignment, target-template alignment, model building, and model evaluation. This unit describes how to calculate comparative models using the program MODELLER and how to use the ModBase database of such models, and discusses all four steps of comparative modeling, frequently observed errors, and some applications. Modeling lactate dehydrogenase from Trichomonas vaginalis (TvLDH) is described as an example. The download and installation of the MODELLER software is also described. © 2016 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Benjamin Webb
- University of California at San Francisco, San Francisco, California
| | - Andrej Sali
- University of California at San Francisco, San Francisco, California
| |
Collapse
|
38
|
Costanzi S, Skorski M, Deplano A, Habermehl B, Mendoza M, Wang K, Biederman M, Dawson J, Gao J. Homology modeling of a Class A GPCR in the inactive conformation: A quantitative analysis of the correlation between model/template sequence identity and model accuracy. J Mol Graph Model 2016; 70:140-152. [PMID: 27723562 DOI: 10.1016/j.jmgm.2016.10.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 09/12/2016] [Accepted: 10/03/2016] [Indexed: 01/22/2023]
Abstract
With the present work we quantitatively studied the modellability of the inactive state of Class A G protein-coupled receptors (GPCRs). Specifically, we constructed models of one of the Class A GPCRs for which structures solved in the inactive state are available, namely the β2 AR, using as templates each of the other class members for which structures solved in the inactive state are also available. Our results showed a detectable linear correlation between model accuracy and model/template sequence identity. This suggests that the likely accuracy of the homology models that can be built for a given receptor can be generally forecasted on the basis of the available templates. We also probed whether sequence alignments that allow for the presence of gaps within the transmembrane domains to account for structural irregularities afford better models than the classical alignment procedures that do not allow for the presence of gaps within such domains. As our results indicated, although the overall differences are very subtle, the inclusion of internal gaps within the transmembrane domains has a noticeable a beneficial effect on the local structural accuracy of the domain in question.
Collapse
Affiliation(s)
- Stefano Costanzi
- Department of Chemistry, American University, Washington, DC 20016, USA; Center for Behavioral Neuroscience, American University, Washington, DC 20016, USA.
| | - Matthew Skorski
- Department of Chemistry, American University, Washington, DC 20016, USA
| | | | - Brett Habermehl
- Department of Chemistry, American University, Washington, DC 20016, USA
| | - Mary Mendoza
- Department of Chemistry, American University, Washington, DC 20016, USA
| | - Keyun Wang
- Department of Chemistry, American University, Washington, DC 20016, USA
| | | | - Jessica Dawson
- Department of Chemistry, American University, Washington, DC 20016, USA
| | - Jia Gao
- Department of Chemistry, American University, Washington, DC 20016, USA
| |
Collapse
|
39
|
Webb B, Sali A. Comparative Protein Structure Modeling Using MODELLER. CURRENT PROTOCOLS IN BIOINFORMATICS 2016; 54:5.6.1-5.6.37. [PMID: 27322406 PMCID: PMC5031415 DOI: 10.1002/cpbi.3] [Citation(s) in RCA: 1943] [Impact Index Per Article: 242.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Comparative protein structure modeling predicts the three-dimensional structure of a given protein sequence (target) based primarily on its alignment to one or more proteins of known structure (templates). The prediction process consists of fold assignment, target-template alignment, model building, and model evaluation. This unit describes how to calculate comparative models using the program MODELLER and how to use the ModBase database of such models, and discusses all four steps of comparative modeling, frequently observed errors, and some applications. Modeling lactate dehydrogenase from Trichomonas vaginalis (TvLDH) is described as an example. The download and installation of the MODELLER software is also described. © 2016 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Benjamin Webb
- University of California at San Francisco, San Francisco, California
| | - Andrej Sali
- University of California at San Francisco, San Francisco, California
| |
Collapse
|
40
|
Shimizu H, Miyazaki H, Ohsawa N, Shoji S, Ishizuka-Katsura Y, Tosaki A, Oyama F, Terada T, Sakamoto K, Shirouzu M, Sekine SI, Nukina N, Yokoyama S. Structure-based site-directed photo-crosslinking analyses of multimeric cell-adhesive interactions of voltage-gated sodium channel β subunits. Sci Rep 2016; 6:26618. [PMID: 27216889 PMCID: PMC4877568 DOI: 10.1038/srep26618] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 05/04/2016] [Indexed: 11/09/2022] Open
Abstract
The β1, β2, and β4 subunits of voltage-gated sodium channels reportedly function as cell adhesion molecules. The present crystallographic analysis of the β4 extracellular domain revealed an antiparallel arrangement of the β4 molecules in the crystal lattice. The interface between the two antiparallel β4 molecules is asymmetric, and results in a multimeric assembly. Structure-based mutagenesis and site-directed photo-crosslinking analyses of the β4-mediated cell-cell adhesion revealed that the interface between the antiparallel β4 molecules corresponds to that in the trans homophilic interaction for the multimeric assembly of β4 in cell-cell adhesion. This trans interaction mode is also employed in the β1-mediated cell-cell adhesion. Moreover, the β1 gene mutations associated with generalized epilepsy with febrile seizures plus (GEFS+) impaired the β1-mediated cell-cell adhesion, which should underlie the GEFS+ pathogenesis. Thus, the structural basis for the β-subunit-mediated cell-cell adhesion has been established.
Collapse
Affiliation(s)
- Hideaki Shimizu
- RIKEN Systems and Structural Biology Center, Tsurumi, Yokohama 230-0045, Japan.,RIKEN Center for Life Science Technologies, Tsurumi, Yokohama 230-0045, Japan.,Laboratory for Structural Neuropathology, RIKEN Brain Science Institute, Wako, Saitama 351-0198, Japan
| | - Haruko Miyazaki
- Laboratory for Structural Neuropathology, RIKEN Brain Science Institute, Wako, Saitama 351-0198, Japan.,Department of Neuroscience for Neurodegenerative Disorders, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan.,Laboratory of Structural Neuropathology, Doshisha University Graduate School of Brain Science, 1-3 Tatara Miyakodani, Kyotanabe-shi, Kyoto 610-0394, Japan
| | - Noboru Ohsawa
- RIKEN Systems and Structural Biology Center, Tsurumi, Yokohama 230-0045, Japan.,RIKEN Center for Life Science Technologies, Tsurumi, Yokohama 230-0045, Japan
| | - Shisako Shoji
- RIKEN Systems and Structural Biology Center, Tsurumi, Yokohama 230-0045, Japan.,RIKEN Center for Life Science Technologies, Tsurumi, Yokohama 230-0045, Japan
| | - Yoshiko Ishizuka-Katsura
- RIKEN Systems and Structural Biology Center, Tsurumi, Yokohama 230-0045, Japan.,RIKEN Center for Life Science Technologies, Tsurumi, Yokohama 230-0045, Japan
| | - Asako Tosaki
- Laboratory for Structural Neuropathology, RIKEN Brain Science Institute, Wako, Saitama 351-0198, Japan
| | - Fumitaka Oyama
- Laboratory for Structural Neuropathology, RIKEN Brain Science Institute, Wako, Saitama 351-0198, Japan.,Department of Chemistry and Life Science, Kogakuin University, Hachioji, Tokyo 192-0015, Japan
| | - Takaho Terada
- RIKEN Systems and Structural Biology Center, Tsurumi, Yokohama 230-0045, Japan.,RIKEN Structural Biology Laboratory, Tsurumi, Yokohama 230-0045, Japan
| | - Kensaku Sakamoto
- RIKEN Systems and Structural Biology Center, Tsurumi, Yokohama 230-0045, Japan.,RIKEN Center for Life Science Technologies, Tsurumi, Yokohama 230-0045, Japan
| | - Mikako Shirouzu
- RIKEN Systems and Structural Biology Center, Tsurumi, Yokohama 230-0045, Japan.,RIKEN Center for Life Science Technologies, Tsurumi, Yokohama 230-0045, Japan
| | - Shun-Ichi Sekine
- RIKEN Systems and Structural Biology Center, Tsurumi, Yokohama 230-0045, Japan.,RIKEN Center for Life Science Technologies, Tsurumi, Yokohama 230-0045, Japan
| | - Nobuyuki Nukina
- Laboratory for Structural Neuropathology, RIKEN Brain Science Institute, Wako, Saitama 351-0198, Japan.,Department of Neuroscience for Neurodegenerative Disorders, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan.,Laboratory of Structural Neuropathology, Doshisha University Graduate School of Brain Science, 1-3 Tatara Miyakodani, Kyotanabe-shi, Kyoto 610-0394, Japan
| | - Shigeyuki Yokoyama
- RIKEN Systems and Structural Biology Center, Tsurumi, Yokohama 230-0045, Japan.,RIKEN Structural Biology Laboratory, Tsurumi, Yokohama 230-0045, Japan
| |
Collapse
|
41
|
Chura-Chambi RM, Arcuri HA, Lino F, Versati N, Palma MS, Favaro DC, Morganti L. Structural studies of the protein endostatin in fusion with BAX BH3 death domain, a hybrid that presents enhanced antitumoral activity. Biotechnol Appl Biochem 2016; 64:356-363. [PMID: 27144384 DOI: 10.1002/bab.1503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 04/28/2016] [Indexed: 11/12/2022]
Abstract
Endostatin (ES) is an antiangiogenic protein that exhibits antitumor activity in animal models. However, the activity observed in animals was not observed in human clinical trials. ES-BAX is a fusion protein composed of two functional domains: ES, which presents specificity and is internalized by activated endothelial cells and the proapoptotic BH3 domain of the protein BAX, a peptide inductor of cellular death when internalized. We have previously shown (Chura-Chambi et al., Cell Death Dis, 5, e1371, 2014) that ES-BAX presents improved antitumor activity in relation to wild-type ES. Secondary and tertiary structures of ES-BAX are similar to ES, as indicated by homology-modeling studies and molecular dynamics simulations. Tryptophan intrinsic fluorescence and circular dichroism spectroscopy corroborate these data. 15 N HSQC NMR indicates that ES-BAX is structured, but some ES residues have suffered chemical shift perturbations, suggesting that the BH3 peptide interacts with some parts of the ES protein. ES and ES-BAX present similar stability to thermal denaturation. The production of stable hybrid proteins can be a new approach to the development of therapeutic agents presenting specificity for tumoral endothelium and improved antitumor effect.
Collapse
Affiliation(s)
- Rosa Maria Chura-Chambi
- Centro de Biotecnologia, Instituto de Pesquisas Energéticas e Nucleares, IPEN-CNEN/SP, São Paulo, Brazil
| | - Helen Andrade Arcuri
- Departamento de Imunologia Clínica e Alergia da Faculdade de Medicina da USP, São Paulo, Brazil
| | - Felipe Lino
- Centro de Biotecnologia, Instituto de Pesquisas Energéticas e Nucleares, IPEN-CNEN/SP, São Paulo, Brazil
| | - Natan Versati
- Centro de Biotecnologia, Instituto de Pesquisas Energéticas e Nucleares, IPEN-CNEN/SP, São Paulo, Brazil
| | - Mario Sergio Palma
- Centro de Estudos de Insetos Sociais, Instituto de Biociências de Rio Claro, UNESP, Rio Claro, São Paulo, Brazil
| | - Denize C Favaro
- Departamento de Química, Instituto de Ciências Exatas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ligia Morganti
- Centro de Biotecnologia, Instituto de Pesquisas Energéticas e Nucleares, IPEN-CNEN/SP, São Paulo, Brazil
| |
Collapse
|
42
|
Khan FI, Wei DQ, Gu KR, Hassan MI, Tabrez S. Current updates on computer aided protein modeling and designing. Int J Biol Macromol 2016; 85:48-62. [DOI: 10.1016/j.ijbiomac.2015.12.072] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 12/17/2015] [Accepted: 12/21/2015] [Indexed: 12/15/2022]
|
43
|
Ding Y, Qiu Y, Zou L, Tan Z, Dai J, Xu W. Three conserved MyD88-recruiting TLR residues exert different effects on the human TLR4 signaling pathway. Immunol Res 2016; 62:213-21. [PMID: 25948473 DOI: 10.1007/s12026-015-8652-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Stimulation of Toll-like receptor (TLR) 4 leads to the activation of both MyD88-dependent and MyD88-independent pathways through the recruitment of adaptors TIRAP/MyD88 and TRIF/TRAM, respectively. However, the molecular basis of the TLR4 Toll/interleukin-1 receptor (TIR) domain in recruiting these downstream adaptors is still not entirely clear. Here, we identify three amino acid residues (714P in the BB loop, 696L in the αA helix and 721N in the αB sheet) conserved in all MyD88-recruited TLRs, but not the TLR3 TIR domain, as being critical for TLR4 responsiveness to LPS. These results were based on the substitution of each residue with a residue of the opposite type (hydrophilic/hydrophobic). However, the responsiveness of the TLR4 mutants to LPS was only partially decreased when each residue was replaced with a residue having the same hydrophilicity/hydrophobicity. This result is likely associated with an alteration in the BB-loop conformation of each TLR4 mutant and its ability to recruit the downstream adaptor TRAM. Thus, we identified three amino acids essential for TLR4 signaling, and their replacement with a residue of the same or opposite hydrophilicity/hydrophobicity greatly affected TLR4 signaling. This study furthers our understanding of the molecular mechanism by which the TLR4 TIR domain modulates TLR4 signaling and also provides new insight for the design of antisepsis therapy.
Collapse
Affiliation(s)
- Yan Ding
- Department of Pathogenic Biology, Third Military Medical University, 30 Gaotanyan Zhengjie, Shapingba District, Chongqing, 400038, People's Republic of China
| | | | | | | | | | | |
Collapse
|
44
|
Kanwal S, Jamil F, Ali A, Sehgal SA. Comparative Modeling, Molecular Docking, and Revealing of Potential Binding Pockets of RASSF2; a Candidate Cancer Gene. Interdiscip Sci 2016; 9:214-223. [PMID: 26782783 DOI: 10.1007/s12539-016-0145-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2015] [Revised: 12/04/2015] [Accepted: 01/06/2016] [Indexed: 12/16/2022]
Abstract
RASSF2, potential tumor suppressor gene, acts as a KRAS-specific effectors protein and may promote apoptosis and cell cycle arrest. It stabilizes STK3/MST2 by protecting it from proteasomal degradation. RASSF2 plays a significant role against the inhibition of cancer. MODELLER (9v15) and online servers (I-Tasser, SwissModel, 3D-JigSaw, ModWeb) were utilized to generate 3D structures of the RASSF2 based on homology modeling. A comparison between models predicted by MODELLER (9v15) and Web servers had been checked through utilized evaluation tools. The most potent model for RASSF2 was analyzed and selected for molecular docking studies. The binding pockets were revealed for binding studies through Site Hound. AutoDock Vina and AutoDock4 were utilized for molecular docking, and the attempt of this experiment was to identify the ligands for RASSF2. The selected compounds may act as regulators and regulate the normal activity of RASSF2. It was also analyzed and observed that the selected compounds showed least binding energy and high-affinity binding in predicted top binding domain. The determination of protein function is based on accurate identification of binding sites in protein structures. The binding site is known, and it may allow the ligand type and protein function to be determined by performing in silico and experimental procedures. The detection, comparison, and analysis of binding pockets are pivotal to drug discovery. It proposed that predicted structure is reliable for the structural insights and functional studies. The predicted binding pockets may lead to further analysis (drug discovery), used against cancer study.
Collapse
Affiliation(s)
- Sonia Kanwal
- Department of Biosciences, COMSATS Institute of Information Technology, Sahiwal, Pakistan
| | - Farrukh Jamil
- Department of Biosciences, COMSATS Institute of Information Technology, Sahiwal, Pakistan
| | - Ahmad Ali
- Department of Biosciences, COMSATS Institute of Information Technology, Sahiwal, Pakistan
| | - Sheikh Arslan Sehgal
- Department of Biosciences, COMSATS Institute of Information Technology, Sahiwal, Pakistan.
- State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
- University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
45
|
Development and Validation of a Small Single-domain Antibody That Effectively Inhibits Matrix Metalloproteinase 8. Mol Ther 2016; 24:890-902. [PMID: 26775809 DOI: 10.1038/mt.2016.2] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 12/31/2015] [Indexed: 12/15/2022] Open
Abstract
A detrimental role for matrix metalloproteinase 8 (MMP8) has been identified in several pathological conditions, e.g., lethal hepatitis and the systemic inflammatory response syndrome. Since matrix MMP8-deficient mice are protected in the above-mentioned diseases, specific MMP8 inhibitors could be of clinical value. However, targeting a specific matrix metalloproteinase remains challenging due to the strong structural homology of matrix metalloproteinases, which form a family of 25 members in mammals. Single-domain antibodies, called nanobodies, offer a range of possibilities toward therapy since they are easy to generate, express, produce, and modify, e.g., by linkage to nanobodies directed against other target molecules. Hence, we generated small MMP8-binding nanobodies, and established a proof-of-principle for developing nanobodies that inhibit matrix metalloproteinase activity. Also, we demonstrated for the first time the possibility of expressing nanobodies systemically by in vivo electroporation of the muscle and its relevance as a potential therapy in inflammatory diseases.
Collapse
|
46
|
Wang H, El Maadidi S, Fischer J, Grabski E, Dickhöfer S, Klimosch S, Flannery SM, Filomena A, Wolz OO, Schneiderhan-Marra N, Löffler MW, Wiese M, Pichulik T, Müllhaupt B, Semela D, Dufour JF, Bochud PY, Bowie AG, Kalinke U, Berg T, Weber ANR. A frequent hypofunctional IRAK2 variant is associated with reduced spontaneous hepatitis C virus clearance. Hepatology 2015; 62:1375-87. [PMID: 26250868 DOI: 10.1002/hep.28105] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Accepted: 08/03/2015] [Indexed: 12/29/2022]
Abstract
UNLABELLED Patients carrying very rare loss-of-function mutations in interleukin-1 receptor-associated kinase 4 (IRAK4), a critical signaling mediator in Toll-like receptor signaling, are severely immunodeficient, highlighting the paramount role of IRAK kinases in innate immunity. We discovered a comparatively frequent coding variant of the enigmatic human IRAK2, L392V (rs3844283), which is found homozygously in ∼15% of Caucasians, to be associated with a reduced ability to induce interferon-alpha in primary human plasmacytoid dendritic cells in response to hepatitis C virus (HCV). Cytokine production in response to purified Toll-like receptor agonists was also impaired. Additionally, rs3844283 was epidemiologically associated with a chronic course of HCV infection in two independent HCV cohorts and emerged as an independent predictor of chronic HCV disease. Mechanistically, IRAK2 L392V showed intact binding to, but impaired ubiquitination of, tumor necrosis factor receptor-associated factor 6, a vital step in signal transduction. CONCLUSION Our study highlights IRAK2 and its genetic variants as critical factors and potentially novel biomarkers for human antiviral innate immunity.
Collapse
Affiliation(s)
- Hui Wang
- Junior Research Group Toll-Like Receptors and Cancer, German Cancer Research Center, Heidelberg, Germany
- Interfaculty Institute for Cell Biology, Department of Immunology, University of Tübingen, Tübingen, Germany
| | - Souhayla El Maadidi
- Interfaculty Institute for Cell Biology, Department of Immunology, University of Tübingen, Tübingen, Germany
| | - Janett Fischer
- Interfaculty Institute for Cell Biology, Department of Immunology, University of Tübingen, Tübingen, Germany
- Section of Hepatology, Clinic for Gastroenterology and Rheumatology, University Hospital Leipzig, Leipzig, Germany
| | - Elena Grabski
- Institute for Experimental Infection Research, TWINCORE Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, Hannover, Germany
| | - Sabine Dickhöfer
- Interfaculty Institute for Cell Biology, Department of Immunology, University of Tübingen, Tübingen, Germany
| | - Sascha Klimosch
- Interfaculty Institute for Cell Biology, Department of Immunology, University of Tübingen, Tübingen, Germany
| | - Sinead M Flannery
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Angela Filomena
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| | - Olaf-Oliver Wolz
- Interfaculty Institute for Cell Biology, Department of Immunology, University of Tübingen, Tübingen, Germany
| | | | - Markus W Löffler
- Interfaculty Institute for Cell Biology, Department of Immunology, University of Tübingen, Tübingen, Germany
- Department of General, Visceral, and Transplant Surgery, University Hospital Tübingen, Tübingen, Germany
| | - Manfred Wiese
- Section of Hepatology, Clinic for Gastroenterology and Rheumatology, University Hospital Leipzig, Leipzig, Germany
| | - Tica Pichulik
- Interfaculty Institute for Cell Biology, Department of Immunology, University of Tübingen, Tübingen, Germany
| | - Beat Müllhaupt
- Gastroenterology and Hepatology Department, University Hospital Zurich, Zurich, Switzerland
| | - David Semela
- Department of Gastroenterology and Hepatology, Canton Hospital St. Gallen, St. Gallen, Switzerland
| | - Jean-François Dufour
- Hepatology Section, Department Visceral Surgery and Medicine, University Hospital Bern, Bern, Switzerland
| | | | - Andrew G Bowie
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Ulrich Kalinke
- Institute for Experimental Infection Research, TWINCORE Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, Hannover, Germany
| | - Thomas Berg
- Section of Hepatology, Clinic for Gastroenterology and Rheumatology, University Hospital Leipzig, Leipzig, Germany
| | - Alexander N R Weber
- Junior Research Group Toll-Like Receptors and Cancer, German Cancer Research Center, Heidelberg, Germany
- Interfaculty Institute for Cell Biology, Department of Immunology, University of Tübingen, Tübingen, Germany
| |
Collapse
|
47
|
Rossetti G, Dibenedetto D, Calandrini V, Giorgetti A, Carloni P. Structural predictions of neurobiologically relevant G-protein coupled receptors and intrinsically disordered proteins. Arch Biochem Biophys 2015; 582:91-100. [DOI: 10.1016/j.abb.2015.03.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Revised: 03/11/2015] [Accepted: 03/12/2015] [Indexed: 01/05/2023]
|
48
|
Paital B, Sablok G, Kumar S, Singh SK, Chainy GBN. Investigating the Conformational Structure and Potential Site Interactions of SOD Inhibitors on Ec-SOD in Marine Mud Crab Scylla serrata: A Molecular Modeling Approach. Interdiscip Sci 2015; 8:312-8. [PMID: 26286009 DOI: 10.1007/s12539-015-0110-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Revised: 01/21/2015] [Accepted: 02/26/2015] [Indexed: 10/23/2022]
Abstract
Superoxide dismutases (SODs) act as a first line of the enzymatic antioxidant defense system to control cellular superoxide anion toxicity. Previously, several inhibitors have been widely identified and catalogued for inhibition of SOD activity; however, still the information about the mechanism of interaction and points toward the inhibitor interactions in structures of SODs in general and in extracellular (Ec)-SOD in particular is still in naive. In the present research, we present an insight to elucidate the molecular basis of interactions of SOD inhibitors with Ec-SOD in mud crab Scylla serrata using molecular modeling and docking approaches. Different inhibitors of SOD such as hydrogen peroxide [Formula: see text], potassium cyanide, sodium dodecyl sulfate (SDS), [Formula: see text]-mercaptoethanol and dithiocarbamate were screened to understand the potential sites that may act as sites for cleavage or blocking in the protein. SOD-SDS and [Formula: see text] complex interactions indicate residues Pro72 and Asp102 of the predicted crab Ec-SOD as common targets. The GOLD result indicates that Pro72, Asp102 and Thr103 are commonly acting as the site of interaction in Ec-SOD of S. serrata with SOD inhibitors. For the first time, the results of this study provide an insight into the structural properties of Ec-SOD of S. serrata and define the possible involvements between the amino acids present in its active sites, i.e., in the regions from 70 to 84 and from 101 to 103 and different inhibitors.
Collapse
Affiliation(s)
- Biswaranjan Paital
- Department of Zoology, College of Basic Science and Humanities, Orissa University of Agriculture and Technology, Bhubaneswar, 751003, Odisha, India.
| | - Gaurav Sablok
- Plant Functional Biology and Climate Change Cluster C3, University of Technology Sydney, PO Box 123, Broadway, NSW, 2007, Australia
| | - Sunil Kumar
- Bioinformatics Centre, Institute of Life Sciences, Nalco Square, Bhubaneswar, 751023, India. .,National Bureau of Agriculturally Important Microorganisms (NBAIM), Indian Council of Agricultural Research (ICAR), Govt. of India, Mau, Uttar Pradesh, 275103, India.
| | - Sanjeev Kumar Singh
- Computer Aided Drug Design and Molecular Modeling Lab, Department of Bioinformatics, Alagappa University, Karaikudi, Tamil Nadu, 630003, India
| | - G B N Chainy
- Department of Biotechnology, Utkal University, Bhubaneswar, India
| |
Collapse
|
49
|
Kallubai M, Amineni U, Mallavarapu M, Kadiyala V. In Silico Approach to Support that p-Nitrophenol Monooxygenase from Arthrobacter sp. Strain JS443 Catalyzes the Initial Two Sequential Monooxygenations. Interdiscip Sci 2015; 7:157-67. [PMID: 26272475 DOI: 10.1007/s12539-015-0018-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Revised: 06/25/2014] [Accepted: 10/29/2014] [Indexed: 10/23/2022]
Abstract
p-Nitrophenol (PNP), used primarily for manufacturing pesticides and dyes, has been recognized as a priority environmental pollutant. It is therefore important to reduce the input of this toxicant into the environment and to establish approaches for its removal from the contaminated sites. PNP monooxygenase, a novel enzyme from Gram-positive bacteria like Arthrobacter sp. and Bacillus sp., that comprises two components, a flavoprotein reductase and an oxygenase, catalyzes the initial two sequential monooxygenations to convert PNP to trihydroxybenzene. Accurate and reliable prediction of this enzyme-substrate interactions and binding affinity are of vital importance in understanding these catalytic mechanisms of the two sequential reactions. As crystal structure of the enzyme has not yet been published, we built a homology model for PNP monooxygenase using crystallized chlorophenol 4-monooxygenase from Burkholderia cepacia AC1100 (3HWC) as the template. The model was assessed for its reliability using PROCHECK, ERRAT and ProSA. Molecular docking of the physiological substrates, PNP and 4-nitrocatechol (4-NC), was carried out using Glide v5.7 implemented in Maestro v9.2, and the binding energies were calculated to substantiate the prediction. Docking complexes formed by molecular level interactions of PNP monooxygenase-PNP/4-NC without or with the cofactors, FAD and NADH, showed good correlation with the established experimental evidence that the two-component PNP monooxygenase catalyzes both the hydroxylation of PNP and the oxidative release of nitrite from 4-NC in B. sphaericus JS905. Furthermore, molecular dynamics simulations performed for docking complexes using Desmond v3.0 showed stable nature of the interactions as well.
Collapse
Affiliation(s)
- Monika Kallubai
- Department of Microbiology, Sri Krishnadevaraya University, Anantapur, 515055, India
| | | | | | | |
Collapse
|
50
|
Cluxton CD, Caffrey BE, Kinsella GK, Moynagh PN, Fares MA, Fallon PG. Functional conservation of an ancestral Pellino protein in helminth species. Sci Rep 2015; 5:11687. [PMID: 26120048 PMCID: PMC4484250 DOI: 10.1038/srep11687] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Accepted: 05/26/2015] [Indexed: 12/27/2022] Open
Abstract
The immune system of H. sapiens has innate signaling pathways that arose in ancestral species. This is exemplified by the discovery of the Toll-like receptor (TLR) pathway using free-living model organisms such as Drosophila melanogaster. The TLR pathway is ubiquitous and controls sensitivity to pathogen-associated molecular patterns (PAMPs) in eukaryotes. There is, however, a marked absence of this pathway from the plathyhelminthes, with the exception of the Pellino protein family, which is present in a number of species from this phylum. Helminth Pellino proteins are conserved having high similarity, both at the sequence and predicted structural protein level, with that of human Pellino proteins. Pellino from a model helminth, Schistosoma mansoni Pellino (SmPellino), was shown to bind and poly-ubiquitinate human IRAK-1, displaying E3 ligase activity consistent with its human counterparts. When transfected into human cells SmPellino is functional, interacting with signaling proteins and modulating mammalian signaling pathways. Strict conservation of a protein family in species lacking its niche signalling pathway is rare and provides a platform to examine the ancestral functions of Pellino proteins that may translate into novel mechanisms of immune regulation in humans.
Collapse
Affiliation(s)
- Christopher D Cluxton
- School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Brian E Caffrey
- Department of Genetics, Trinity College Dublin, Dublin 2, Ireland
| | - Gemma K Kinsella
- Institute of Immunology, National University of Ireland Maynooth, County Kildare, Ireland
| | - Paul N Moynagh
- Institute of Immunology, National University of Ireland Maynooth, County Kildare, Ireland
| | - Mario A Fares
- 1] Department of Genetics, Trinity College Dublin, Dublin 2, Ireland [2] Integrative Systems Biology Group, Instituto de Biología Molecular y Celular de Plantas (C.S.I.C-UPV)
| | - Padraic G Fallon
- 1] School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland [2] National Children's Research Centre, Our Lady's Children's Hospital, Dublin 8, Ireland
| |
Collapse
|