Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
2.
Sci Rep ; 13(1): 13668, 2023 08 22.
Article in English | MEDLINE | ID: mdl-37608223

ABSTRACT

Coronaviruses have been the causative agent of three epidemics and pandemics in the past two decades, including the ongoing COVID-19 pandemic. A broadly-neutralizing coronavirus therapeutic is desirable not only to prevent and treat COVID-19, but also to provide protection for high-risk populations against future emergent coronaviruses. As all coronaviruses use spike proteins on the viral surface to enter the host cells, and these spike proteins share sequence and structural homology, we set out to discover cross-reactive biologic agents targeting the spike protein to block viral entry. Through llama immunization campaigns, we have identified single domain antibodies (VHHs) that are cross-reactive against multiple emergent coronaviruses (SARS-CoV, SARS-CoV-2, and MERS). Importantly, a number of these antibodies show sub-nanomolar potency towards all SARS-like viruses including emergent CoV-2 variants. We identified nine distinct epitopes on the spike protein targeted by these VHHs. Further, by engineering VHHs targeting distinct, conserved epitopes into multi-valent formats, we significantly enhanced their neutralization potencies compared to the corresponding VHH cocktails. We believe this approach is ideally suited to address both emerging SARS-CoV-2 variants during the current pandemic as well as potential future pandemics caused by SARS-like coronaviruses.


Subject(s)
COVID-19 , Camelids, New World , Single-Domain Antibodies , Humans , Animals , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Pandemics , Epitopes
3.
Front Immunol ; 13: 864775, 2022.
Article in English | MEDLINE | ID: mdl-35603164

ABSTRACT

The SARS-CoV-2 pandemic and particularly the emerging variants have deepened the need for widely available therapeutic options. We have demonstrated that hexamer-enhancing mutations in the Fc region of anti-SARS-CoV IgG antibodies lead to a noticeable improvement in IC50 in both pseudo and live virus neutralization assay compared to parental molecules. We also show that hexamer-enhancing mutants improve C1q binding to target surface. To our knowledge, this is the first time this format has been explored for application in viral neutralization and the studies provide proof-of-concept for the use of hexamer-enhanced IgG1 molecules as potential anti-viral therapeutics.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , Immunoglobulin G/genetics , Immunologic Tests , Pandemics , SARS-CoV-2/genetics
4.
J Med Chem ; 65(7): 5593-5605, 2022 04 14.
Article in English | MEDLINE | ID: mdl-35298158

ABSTRACT

We have identified a series of novel insulin receptor partial agonists (IRPAs) with a potential to mitigate the risk of hypoglycemia associated with the use of insulin as an antidiabetic treatment. These molecules were designed as dimers of native insulin connected via chemical linkers of variable lengths with optional capping groups at the N-terminals of insulin chains. Depending on the structure, the maximal activation level (%Max) varied in the range of ∼20-70% of native insulin, and EC50 values remained in sub-nM range. Studies in minipig and dog demonstrated that IRPAs had sufficient efficacy to normalize plasma glucose levels in diabetes, while providing reduction of hypoglycemia risk. IRPAs had a prolonged duration of action, potentially making them suitable for once-daily dosing. Two lead compounds with %Max values of 30 and 40% relative to native insulin were selected for follow up studies in the clinic.


Subject(s)
Diabetes Mellitus, Type 2 , Hypoglycemia , Animals , Blood Glucose , Diabetes Mellitus, Type 2/drug therapy , Dogs , Hypoglycemia/drug therapy , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Insulin/therapeutic use , Receptor, Insulin , Swine , Swine, Miniature , Therapeutic Index
5.
Nat Commun ; 13(1): 942, 2022 02 17.
Article in English | MEDLINE | ID: mdl-35177603

ABSTRACT

Insulin analogs have been developed to treat diabetes with focus primarily on improving the time action profile without affecting ligand-receptor interaction or functional selectivity. As a result, inherent liabilities (e.g. hypoglycemia) of injectable insulin continue to limit the true therapeutic potential of related agents. Insulin dimers were synthesized to investigate whether partial agonism of the insulin receptor (IR) tyrosine kinase is achievable, and to explore the potential for tissue-selective systemic insulin pharmacology. The insulin dimers induced distinct IR conformational changes compared to native monomeric insulin and substrate phosphorylation assays demonstrated partial agonism. Structurally distinct dimers with differences in conjugation sites and linkers were prepared to deliver desirable IR partial agonist (IRPA). Systemic infusions of a B29-B29 dimer in vivo revealed sharp differences compared to native insulin. Suppression of hepatic glucose production and lipolysis were like that attained with regular insulin, albeit with a distinctly shallower dose-response. In contrast, there was highly attenuated stimulation of glucose uptake into muscle. Mechanistic studies indicated that IRPAs exploit tissue differences in receptor density and have additional distinctions pertaining to drug clearance and distribution. The hepato-adipose selective action of IRPAs is a potentially safer approach for treatment of diabetes.


Subject(s)
Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 1/drug therapy , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Receptor, Insulin/agonists , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Alloxan/administration & dosage , Alloxan/toxicity , Animals , Blood Glucose/drug effects , Blood Glucose/metabolism , CHO Cells , Cricetulus , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/chemically induced , Diabetes Mellitus, Type 1/metabolism , HEK293 Cells , Humans , Hypoglycemic Agents/therapeutic use , Insulin/therapeutic use , Lipolysis/drug effects , Liver/drug effects , Liver/metabolism , Male , Mice , Rats , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Signal Transduction/drug effects , Swine , Swine, Miniature
6.
Proc Natl Acad Sci U S A ; 118(51)2021 12 21.
Article in English | MEDLINE | ID: mdl-34921117

ABSTRACT

Over the past five decades, tremendous effort has been devoted to computational methods for predicting properties of ligands-i.e., molecules that bind macromolecular targets. Such methods, which are critical to rational drug design, fall into two categories: physics-based methods, which directly model ligand interactions with the target given the target's three-dimensional (3D) structure, and ligand-based methods, which predict ligand properties given experimental measurements for similar ligands. Here, we present a rigorous statistical framework to combine these two sources of information. We develop a method to predict a ligand's pose-the 3D structure of the ligand bound to its target-that leverages a widely available source of information: a list of other ligands that are known to bind the same target but for which no 3D structure is available. This combination of physics-based and ligand-based modeling improves pose prediction accuracy across all major families of drug targets. Using the same framework, we develop a method for virtual screening of drug candidates, which outperforms standard physics-based and ligand-based virtual screening methods. Our results suggest broad opportunities to improve prediction of various ligand properties by combining diverse sources of information through customized machine-learning approaches.


Subject(s)
Antipsychotic Agents/chemistry , Antipsychotic Agents/pharmacology , Drug Design/methods , Artificial Intelligence , Binding Sites , Gene Expression Regulation/drug effects , Ligands , Molecular Docking Simulation , Molecular Structure , Protein Binding , Protein Conformation , Receptors, Dopamine D2/chemistry , Receptors, Dopamine D2/metabolism , Structure-Activity Relationship
7.
J Chem Inf Model ; 61(7): 3696-3707, 2021 07 26.
Article in English | MEDLINE | ID: mdl-34251810

ABSTRACT

Biased agonists, which selectively stimulate certain signaling pathways controlled by a G protein-coupled receptor (GPCR), hold great promise as drugs that maximize efficacy while minimizing dangerous side effects. Biased agonists of the µ-opioid receptor (µOR) are of particular interest as a means to achieve analgesia through G protein signaling without dose-limiting side effects such as respiratory depression and constipation. Rational structure-based design of biased agonists remains highly challenging, however, because the ligand-mediated interactions that are key to activation of each signaling pathway remain unclear. We identify several compounds for which the R- and S-enantiomers have distinct bias profiles at the µOR. These compounds serve as excellent comparative tools to study bias because the identical physicochemical properties of enantiomer pairs ensure that differences in bias profiles are due to differences in interactions with the µOR binding pocket. Atomic-level simulations of compounds at µOR indicate that R- and S-enantiomers adopt different poses that form distinct interactions with the binding pocket. A handful of specific interactions with highly conserved binding pocket residues appear to be responsible for substantial differences in arrestin recruitment between enantiomers. Our results offer guidance for rational design of biased agonists at µOR and possibly at related GPCRs.


Subject(s)
Receptors, Opioid, mu , Signal Transduction , GTP-Binding Proteins , Humans , Ligands , Pain , Protein Binding , Receptors, Opioid, mu/metabolism
8.
Nat Commun ; 12(1): 815, 2021 02 05.
Article in English | MEDLINE | ID: mdl-33547286

ABSTRACT

Narcolepsy type 1 (NT1) is a chronic neurological disorder that impairs the brain's ability to control sleep-wake cycles. Current therapies are limited to the management of symptoms with modest effectiveness and substantial adverse effects. Agonists of the orexin receptor 2 (OX2R) have shown promise as novel therapeutics that directly target the pathophysiology of the disease. However, identification of drug-like OX2R agonists has proven difficult. Here we report cryo-electron microscopy structures of active-state OX2R bound to an endogenous peptide agonist and a small-molecule agonist. The extended carboxy-terminal segment of the peptide reaches into the core of OX2R to stabilize an active conformation, while the small-molecule agonist binds deep inside the orthosteric pocket, making similar key interactions. Comparison with antagonist-bound OX2R suggests a molecular mechanism that rationalizes both receptor activation and inhibition. Our results enable structure-based discovery of therapeutic orexin agonists for the treatment of NT1 and other hypersomnia disorders.


Subject(s)
Aminopyridines/chemistry , Azepines/chemistry , Orexin Receptor Antagonists/chemistry , Orexin Receptors/chemistry , Peptides/chemistry , Sleep Aids, Pharmaceutical/chemistry , Sulfonamides/chemistry , Triazoles/chemistry , Aminopyridines/metabolism , Azepines/metabolism , Binding Sites , Cloning, Molecular , Cryoelectron Microscopy , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , HEK293 Cells , Humans , Molecular Dynamics Simulation , Orexin Receptor Antagonists/metabolism , Orexin Receptors/agonists , Orexin Receptors/metabolism , Peptides/metabolism , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sleep Aids, Pharmaceutical/metabolism , Sulfonamides/metabolism , Triazoles/metabolism
9.
Sci Rep ; 11(1): 2118, 2021 01 22.
Article in English | MEDLINE | ID: mdl-33483531

ABSTRACT

Lung fibrosis, or the scarring of the lung, is a devastating disease with huge unmet medical need. There are limited treatment options and its prognosis is worse than most types of cancer. We previously discovered that MK-0429 is an equipotent pan-inhibitor of αv integrins that reduces proteinuria and kidney fibrosis in a preclinical model. In the present study, we further demonstrated that MK-0429 significantly inhibits fibrosis progression in a bleomycin-induced lung injury model. In search of newer integrin inhibitors for fibrosis, we characterized monoclonal antibodies discovered using Adimab's yeast display platform. We identified several potent neutralizing integrin antibodies with unique human and mouse cross-reactivity. Among these, Ab-31 blocked the binding of multiple αv integrins to their ligands with IC50s comparable to those of MK-0429. Furthermore, both MK-0429 and Ab-31 suppressed integrin-mediated cell adhesion and latent TGFß activation. In IPF patient lung fibroblasts, TGFß treatment induced profound αSMA expression in phenotypic imaging assays and Ab-31 demonstrated potent in vitro activity at inhibiting αSMA expression, suggesting that the integrin antibody is able to modulate TGFß action though mechanisms beyond the inhibition of latent TGFß activation. Together, our results highlight the potential to develop newer integrin therapeutics for the treatment of fibrotic lung diseases.


Subject(s)
Antibodies/metabolism , Fibroblasts/metabolism , Integrin alphaV/metabolism , Pulmonary Fibrosis/metabolism , Animals , Antibodies/immunology , Bleomycin , CHO Cells , Cells, Cultured , Cricetinae , Cricetulus , Fibroblasts/cytology , Humans , Integrin alphaV/immunology , Male , Mice, Inbred C57BL , Naphthyridines/pharmacology , Propionates/pharmacology , Protein Binding , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/prevention & control
10.
Cell ; 183(7): 1813-1825.e18, 2020 12 23.
Article in English | MEDLINE | ID: mdl-33296703

ABSTRACT

Binding of arrestin to phosphorylated G-protein-coupled receptors (GPCRs) controls many aspects of cell signaling. The number and arrangement of phosphates may vary substantially for a given GPCR, and different phosphorylation patterns trigger different arrestin-mediated effects. Here, we determine how GPCR phosphorylation influences arrestin behavior by using atomic-level simulations and site-directed spectroscopy to reveal the effects of phosphorylation patterns on arrestin binding and conformation. We find that patterns favoring binding differ from those favoring activation-associated conformational change. Both binding and conformation depend more on arrangement of phosphates than on their total number, with phosphorylation at different positions sometimes exerting opposite effects. Phosphorylation patterns selectively favor a wide variety of arrestin conformations, differently affecting arrestin sites implicated in scaffolding distinct signaling proteins. We also reveal molecular mechanisms of these phenomena. Our work reveals the structural basis for the long-standing "barcode" hypothesis and has important implications for design of functionally selective GPCR-targeted drugs.


Subject(s)
Arrestin/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Arrestin/chemistry , Computer Simulation , HEK293 Cells , Humans , Phosphates/metabolism , Phosphopeptides/metabolism , Phosphorylation , Protein Binding , Protein Conformation , Spectrum Analysis
11.
Expert Opin Ther Pat ; 30(11): 825-845, 2020 Nov.
Article in English | MEDLINE | ID: mdl-33052748

ABSTRACT

INTRODUCTION: Toll-like receptors 7 and 8 (TLR7 and TLR8) are endosomal immune receptors that initiate an innate immune response and can facilitate activation of the adaptive immune system. Both preclinical and clinical studies have shown the downstream inflammatory response from TLR7 and TLR8 agonism results in preliminary efficacy for the treatment of cancer, viral infections, and for use as a vaccine adjuvant. AREAS COVERED: This patent review covers recent developments in small molecule TLR7 and TLR8 agonists published between January 2014 - February 2020. We summarize relevant chemical scaffolds, observed structure-activity relationships, and where available, preliminary animal models, and clinical data. EXPERT OPINION: In the last 6 years, there has been significant progress in the optimization of novel TLR7 and TLR8 small molecule agonists. These novel compounds are currently being evaluated in the clinic for multiple antiviral and oncology indications. Clinical data from these trials will provide a clearer outlook on 1) the TLR7/8 engagement necessary to obtain the desired immune response, 2) safety margin improvement using directed delivery, and 3) potential synergistic effects with checkpoint inhibitor combination therapies.


Subject(s)
Immunity, Innate/drug effects , Toll-Like Receptor 7/agonists , Toll-Like Receptor 8/agonists , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/pharmacology , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Antiviral Agents/administration & dosage , Antiviral Agents/pharmacology , Drug Development , Humans , Neoplasms/drug therapy , Neoplasms/immunology , Patents as Topic , Virus Diseases/drug therapy , Virus Diseases/immunology
12.
Nature ; 572(7770): 488-492, 2019 08.
Article in English | MEDLINE | ID: mdl-31367042

ABSTRACT

Cation-chloride cotransporters (CCCs) mediate the electroneutral transport of chloride, potassium and/or sodium across the membrane. They have critical roles in regulating cell volume, controlling ion absorption and secretion across epithelia, and maintaining intracellular chloride homeostasis. These transporters are primary targets for some of the most commonly prescribed drugs. Here we determined the cryo-electron microscopy structure of the Na-K-Cl cotransporter NKCC1, an extensively studied member of the CCC family, from Danio rerio. The structure defines the architecture of this protein family and reveals how cytosolic and transmembrane domains are strategically positioned for communication. Structural analyses, functional characterizations and computational studies reveal the ion-translocation pathway, ion-binding sites and key residues for transport activity. These results provide insights into ion selectivity, coupling and translocation, and establish a framework for understanding the physiological functions of CCCs and interpreting disease-related mutations.


Subject(s)
Cryoelectron Microscopy , Solute Carrier Family 12, Member 2/metabolism , Solute Carrier Family 12, Member 2/ultrastructure , Zebrafish , Amino Acid Sequence , Animals , Binding Sites , Cations, Monovalent/metabolism , Chlorides/metabolism , Cytosol/metabolism , Gitelman Syndrome/genetics , Humans , Ion Transport , Models, Molecular , Molecular Dynamics Simulation , Potassium/metabolism , Protein Domains , Sodium/metabolism , Solute Carrier Family 12, Member 2/chemistry , Solute Carrier Family 12, Member 2/genetics , Zebrafish/genetics
13.
Nat Commun ; 10(1): 3289, 2019 07 23.
Article in English | MEDLINE | ID: mdl-31337749

ABSTRACT

Allosteric modulators are highly desirable as drugs, particularly for G-protein-coupled receptor (GPCR) targets, because allosteric drugs can achieve selectivity between closely related receptors. The mechanisms by which allosteric modulators achieve selectivity remain elusive, however, particularly given recent structures that reveal similar allosteric binding sites across receptors. Here we show that positive allosteric modulators (PAMs) of the M1 muscarinic acetylcholine receptor (mAChR) achieve exquisite selectivity by occupying a dynamic pocket absent in existing crystal structures. This cryptic pocket forms far more frequently in molecular dynamics simulations of the M1 mAChR than in those of other mAChRs. These observations reconcile mutagenesis data that previously appeared contradictory. Further mutagenesis experiments validate our prediction that preventing cryptic pocket opening decreases the affinity of M1-selective PAMs. Our findings suggest opportunities for the design of subtype-specific drugs exploiting cryptic pockets that open in certain receptors but not in other receptors with nearly identical static structures.


Subject(s)
Receptor, Muscarinic M1/chemistry , Receptors, G-Protein-Coupled/chemistry , Allosteric Regulation , Allosteric Site , Crystallography, X-Ray , Drug Design , Ligands , Molecular Dynamics Simulation , Mutagenesis, Site-Directed
14.
Cell ; 176(3): 448-458.e12, 2019 01 24.
Article in English | MEDLINE | ID: mdl-30639101

ABSTRACT

Cannabis elicits its mood-enhancing and analgesic effects through the cannabinoid receptor 1 (CB1), a G protein-coupled receptor (GPCR) that signals primarily through the adenylyl cyclase-inhibiting heterotrimeric G protein Gi. Activation of CB1-Gi signaling pathways holds potential for treating a number of neurological disorders and is thus crucial to understand the mechanism of Gi activation by CB1. Here, we present the structure of the CB1-Gi signaling complex bound to the highly potent agonist MDMB-Fubinaca (FUB), a recently emerged illicit synthetic cannabinoid infused in street drugs that have been associated with numerous overdoses and fatalities. The structure illustrates how FUB stabilizes the receptor in an active state to facilitate nucleotide exchange in Gi. The results compose the structural framework to explain CB1 activation by different classes of ligands and provide insights into the G protein coupling and selectivity mechanisms adopted by the receptor.


Subject(s)
Receptor, Cannabinoid, CB1/metabolism , Receptor, Cannabinoid, CB1/ultrastructure , Animals , Cannabinoid Receptor Agonists/pharmacology , Cannabinoids/pharmacology , Cryoelectron Microscopy/methods , Heterotrimeric GTP-Binding Proteins/metabolism , Humans , Indazoles/pharmacology , Ligands , Protein Binding , Receptor, Cannabinoid, CB1/chemistry , Receptors, Cannabinoid/chemistry , Receptors, Cannabinoid/metabolism , Receptors, Cannabinoid/ultrastructure , Receptors, G-Protein-Coupled/metabolism , Sf9 Cells , Signal Transduction/drug effects
15.
Neuron ; 99(6): 1129-1143, 2018 09 19.
Article in English | MEDLINE | ID: mdl-30236283

ABSTRACT

The impact of molecular dynamics (MD) simulations in molecular biology and drug discovery has expanded dramatically in recent years. These simulations capture the behavior of proteins and other biomolecules in full atomic detail and at very fine temporal resolution. Major improvements in simulation speed, accuracy, and accessibility, together with the proliferation of experimental structural data, have increased the appeal of biomolecular simulation to experimentalists-a trend particularly noticeable in, although certainly not limited to, neuroscience. Simulations have proven valuable in deciphering functional mechanisms of proteins and other biomolecules, in uncovering the structural basis for disease, and in the design and optimization of small molecules, peptides, and proteins. Here we describe, in practical terms, the types of information MD simulations can provide and the ways in which they typically motivate further experimental work.


Subject(s)
Computational Biology , Molecular Dynamics Simulation , Protein Conformation , Proteins/chemistry , Humans , Molecular Biology/methods , Peptides/analysis , Proteins/metabolism
16.
Nature ; 557(7705): 452-456, 2018 05.
Article in English | MEDLINE | ID: mdl-29720655

ABSTRACT

Despite intense interest in discovering drugs that cause G-protein-coupled receptors (GPCRs) to selectively stimulate or block arrestin signalling, the structural mechanism of receptor-mediated arrestin activation remains unclear1,2. Here we reveal this mechanism through extensive atomic-level simulations of arrestin. We find that the receptor's transmembrane core and cytoplasmic tail-which bind distinct surfaces on arrestin-can each independently stimulate arrestin activation. We confirm this unanticipated role of the receptor core, and the allosteric coupling between these distant surfaces of arrestin, using site-directed fluorescence spectroscopy. The effect of the receptor core on arrestin conformation is mediated primarily by interactions of the intracellular loops of the receptor with the arrestin body, rather than the marked finger-loop rearrangement that is observed upon receptor binding. In the absence of a receptor, arrestin frequently adopts active conformations when its own C-terminal tail is disengaged, which may explain why certain arrestins remain active long after receptor dissociation. Our results, which suggest that diverse receptor binding modes can activate arrestin, provide a structural foundation for the design of functionally selective ('biased') GPCR-targeted ligands with desired effects on arrestin signalling.


Subject(s)
Arrestins/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Arrestins/chemistry , Cattle , Ligands , Receptors, G-Protein-Coupled/chemistry , Signal Transduction , Spectrometry, Fluorescence
17.
Proc Natl Acad Sci U S A ; 115(10): 2383-2388, 2018 03 06.
Article in English | MEDLINE | ID: mdl-29463720

ABSTRACT

More than two decades ago, the activation mechanism for the membrane-bound photoreceptor and prototypical G protein-coupled receptor (GPCR) rhodopsin was uncovered. Upon light-induced changes in ligand-receptor interaction, movement of specific transmembrane helices within the receptor opens a crevice at the cytoplasmic surface, allowing for coupling of heterotrimeric guanine nucleotide-binding proteins (G proteins). The general features of this activation mechanism are conserved across the GPCR superfamily. Nevertheless, GPCRs have selectivity for distinct G-protein family members, but the mechanism of selectivity remains elusive. Structures of GPCRs in complex with the stimulatory G protein, Gs, and an accessory nanobody to stabilize the complex have been reported, providing information on the intermolecular interactions. However, to reveal the structural selectivity filters, it will be necessary to determine GPCR-G protein structures involving other G-protein subtypes. In addition, it is important to obtain structures in the absence of a nanobody that may influence the structure. Here, we present a model for a rhodopsin-G protein complex derived from intermolecular distance constraints between the activated receptor and the inhibitory G protein, Gi, using electron paramagnetic resonance spectroscopy and spin-labeling methodologies. Molecular dynamics simulations demonstrated the overall stability of the modeled complex. In the rhodopsin-Gi complex, Gi engages rhodopsin in a manner distinct from previous GPCR-Gs structures, providing insight into specificity determinants.


Subject(s)
Heterotrimeric GTP-Binding Proteins , Rhodopsin , Animals , Cattle , Heterotrimeric GTP-Binding Proteins/chemistry , Heterotrimeric GTP-Binding Proteins/genetics , Heterotrimeric GTP-Binding Proteins/metabolism , Molecular Dynamics Simulation , Mutation , Protein Binding , Protein Conformation , Rhodopsin/chemistry , Rhodopsin/genetics , Rhodopsin/metabolism , Spectrum Analysis
18.
J Chem Inf Model ; 57(9): 2344-2350, 2017 09 25.
Article in English | MEDLINE | ID: mdl-28841378

ABSTRACT

Leishmania major peroxidase (LmP) is structurally and functionally similar to the well-studied yeast Cytochrome c peroxidase (CCP). A recent Brownian dynamics study showed that L. major Cytochrome c (LmCytc) associates with LmP by forming an initial complex with the N-terminal helix A of LmP, followed by a movement toward the electron transfer (ET) site observed in the LmP-LmCytc crystal structure. Critical to forming the active electron transfer complex is an intermolecular Arg-Asp ion pair at the center of the interface. If the dissociation reaction is effectively the reverse of the association reaction, then rupture of the Asp-Arg ion pair should be followed by movement of LmCytc back toward LmP helix A. To test this possibility, we have performed multiple molecular dynamics (MD) simulations of the LmP-LmCytc complex. In five separate simulations, LmCytc is observed to indeed move toward helix A, and in two of the simulations, the Asp-Arg ion pair breaks, which frees LmCytc to fully associate with the LmP helix A secondary binding site. These results support the "bind and crawl" or "velcro" mechanism of association, wherein LmCytc forms a nonspecific electrostatic complex with LmP helix A, followed by a "crawl" toward the ET-active site, where the Asp-Arg ion pair holds the LmCytc in position for rapid ET. These simulations also point to Tyr134LmP as being important in the association/dissociation reactions. Experimentally mutating Tyr134 to Phe was found to decrease Km by 3.6-fold, which is consistent with its predicted role in complex formation by MD simulations.


Subject(s)
Cytochrome-c Peroxidase/chemistry , Cytochrome-c Peroxidase/metabolism , Leishmania major/enzymology , Molecular Dynamics Simulation , Peroxidase/chemistry , Peroxidase/metabolism , Heme/metabolism , Mutation , Oxidation-Reduction , Peroxidase/genetics , Protein Conformation
19.
Proc Natl Acad Sci U S A ; 113(31): 8723-8, 2016 08 02.
Article in English | MEDLINE | ID: mdl-27439869

ABSTRACT

The heme iron of cytochromes P450 must be reduced to bind and activate molecular oxygen for substrate oxidation. Reducing equivalents are derived from a redox partner, which requires the formation of a protein-protein complex. A subject of increasing discussion is the role that redox partner binding plays, if any, in favoring significant structural changes in the P450s that are required for activity. Many P450s now have been shown to experience large open and closed motions. Several structural and spectral studies indicate that the well-studied P450cam adopts the open conformation when its redox partner, putidaredoxin (Pdx), binds, whereas recent NMR studies indicate that this view is incorrect. Given the relevance of this discrepancy to P450 chemistry, it is important to determine whether Pdx favors the open or closed form of P450cam. Here, we have used both computational and experimental isothermal titration calorimetry studies that unequivocally show Pdx favors binding to the open form of P450cam. Analyses of molecular-dynamic trajectories also provide insights into intermediate conformational states that could be relevant to catalysis.


Subject(s)
Bacterial Proteins/chemistry , Cytochrome P-450 Enzyme System/chemistry , Molecular Dynamics Simulation , Protein Conformation , Bacterial Proteins/metabolism , Calorimetry/methods , Camphor 5-Monooxygenase/chemistry , Camphor 5-Monooxygenase/metabolism , Cytochrome P-450 Enzyme System/metabolism , Ferredoxins/chemistry , Ferredoxins/metabolism , Nuclear Magnetic Resonance, Biomolecular , Oxidation-Reduction , Protein Binding , Pseudomonas putida/metabolism , Thermodynamics
20.
Protein Sci ; 25(9): 1757-62, 2016 09.
Article in English | MEDLINE | ID: mdl-27342939

ABSTRACT

The Ramachandran plot distributions of nonglycine residues from experimentally determined structures are routinely described as grouping into one of six major basins: ß, PII , α, αL , ξ and γ'. Recent work describing the most common conformations adopted by pairs of residues in folded proteins [i.e., (φ,ψ)2 -motifs] showed that commonly described major basins are not true single thermodynamic basins, but are composed of distinct subregions that are associated with various conformations of either the preceding or following neighbor residue. Here, as documentation of the extent to which the conformational preferences of a central residue are influenced by the conformations of its two neighbors, we present a set of φ,ψ-plots that are delimited simultaneously by the φ,ψ-angles of its neighboring residues on both sides. The level of influence seen here is typically greater than the influence associated with considering the identities of neighboring residues, implying that the use of this heretofore untapped information can improve the accuracy of structure prediction algorithms and low resolution protein structure refinement.


Subject(s)
Models, Molecular , Protein Folding , Proteins/chemistry , Amino Acid Motifs
SELECTION OF CITATIONS
SEARCH DETAIL
...