Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 97
Filter
Add more filters










Publication year range
1.
JACS Au ; 4(2): 837-846, 2024 Feb 26.
Article in English | MEDLINE | ID: mdl-38425926

ABSTRACT

Fully understanding the mechanism of allosteric regulation in biomolecules requires separating and examining all of the involved factors. In enzyme catalysis, allosteric effector binding shifts the structure and dynamics of the active site, leading to modified energetic (e.g., energy barrier) and dynamical (e.g., diffusion coefficient) factors underlying the catalyzed reaction rate. Such modifications can be subtle and dependent on the type of allosteric effector, representing a fine-tuning of protein function. The microscopic description of allosteric regulation at the level of function-dictating factors has prospective applications in fundamental and pharmaceutical sciences, which is, however, largely missing so far. Here, we characterize the allosteric fine-tuning of enzyme catalysis, using human Pin1 as an example, by performing more than half-millisecond all-atom molecular dynamics simulations. Changes of reaction kinetics and the dictating factors, including the free energy surface along the reaction coordinate and the diffusion coefficient of the reaction dynamics, under various enzyme and allosteric effector binding conditions are examined. Our results suggest equal importance of the energetic and dynamical factors, both of which can be modulated allosterically, and the combined effect determines the final allosteric output. We also reveal the potential dynamic basis for allosteric modulation using an advanced statistical technique to detect function-related conformational dynamics. Methods developed in this work can be applied to other allosteric systems.

2.
J Chem Inf Model ; 63(12): 3892-3902, 2023 06 26.
Article in English | MEDLINE | ID: mdl-37285207

ABSTRACT

Drug resistance in antiviral treatments is a serious public health problem. Viral proteins mutate very fast, giving them a way to escape drugs by lowering drug binding affinity but with compromised function. Human immunodeficiency virus type I (HIV-1) protease, a critical antiretroviral therapeutic target, represents a model for such viral regulation under inhibition. Drug inhibitors of HIV-1 protease lose effectiveness as the protein evolves through several variants to become more resistant. However, the detailed mechanism of drug resistance in HIV-1 protease is still unclear. Here, we test the hypothesis that mutations throughout the protease alter the protein conformational ensemble to weaken protein-inhibitor binding, resulting in an inefficient protease but still viable virus. Comparing conformational ensembles between variants and the wild type helps detect these function-related dynamical changes. All analyses of over 30 µs simulations converge to the conclusion that conformational dynamics of more drug-resistant variants are more different from that of the wild type. Distinct roles of mutations during viral evolution are discussed, including a mutation predominantly contributing to the increase of drug resistance and a mutation that is responsible (synergistically) for restoring catalytic efficiency. Drug resistance is mainly due to altered flap dynamics that hinder the access to the active site. The mutant variant showing the highest drug resistance has the most ″collapsed″ active-site pocket and hence the largest magnitude of hindrance of drug binding. An enhanced difference contact network community analysis is applied to understand allosteric communications. The method summarizes multiple conformational ensembles in one community network and can be used in future studies to detect function-related dynamics in proteins.


Subject(s)
HIV Protease Inhibitors , Humans , HIV Protease Inhibitors/chemistry , Binding Sites , Drug Resistance, Viral/genetics , Catalytic Domain , Mutation , HIV Protease/metabolism
3.
J Phys Chem B ; 127(14): 3139-3150, 2023 04 13.
Article in English | MEDLINE | ID: mdl-36989346

ABSTRACT

Cyclophilins are ubiquitous human enzymes that catalyze peptidyl-prolyl cis-trans isomerization in protein substrates. Of the 17 unique isoforms, five closely related isoforms (CypA-E) are found in various environments and participate in diverse cellular processes, yet all have similar structures and the same core catalytic function. The question is what key residues are behind the conserved function of these enzymes. Here, conformational dynamics are compared across these isoforms to detect conserved dynamics essential for the catalytic activity of cyclophilins. A set of key dynamic residues, defined by the most dynamically conserved positions, are identified in the gatekeeper 2 region. The highly conserved glycine (Gly80) in this region is predicted to underlie the local flexibility, which is further tested by molecular dynamics simulations performed on mutants (G80A) of CypE and CypA. The mutation leads to decreased flexibility of CypE and CypA during substrate binding but increased flexibility during catalysis. Dynamical changes occur in the mutated region and a distal loop downstream of the mutation site in sequence. Examinations of the mutational effect on catalysis show that both mutated CypE and CypA exhibit shifted binding free energies of the substrate under distinct isomer conformations. The results suggest a loss of function in the mutated CypE and CypA. These catalytic changes by the mutation are likely independent of the substrate sequence, at least in CypA. Our work presents a method to identify function-related key residues in proteins.


Subject(s)
Cyclophilins , Proteins , Humans , Protein Conformation , Molecular Dynamics Simulation , Catalysis
4.
Comput Biol Chem ; 104: 107847, 2023 Jun.
Article in English | MEDLINE | ID: mdl-36925366
5.
Biochemistry ; 62(5): 1070-1081, 2023 03 07.
Article in English | MEDLINE | ID: mdl-36795942

ABSTRACT

Numerous studies demonstrate that enzymes undergo multiple conformational changes during catalysis. The malleability of enzymes forms the basis for allosteric regulation: residues located far from the active site can exert long-range dynamical effects on the active site residues to modulate catalysis. The structure of Pseudomonas aeruginosa d-arginine dehydrogenase (PaDADH) shows four loops (L1, L2, L3, and L4) that span the substrate and the FAD-binding domains. Loop L4 comprises residues 329-336, spanning over the flavin cofactor. The I335 residue on loop L4 is ∼10 Šaway from the active site and ∼3.8 Šfrom N(1)-C(2)═O atoms of the flavin. In this study, we used molecular dynamics and biochemical techniques to investigate the effect of the mutation of I335 to histidine on the catalytic function of PaDADH. Molecular dynamics showed that the conformational dynamics of PaDADH are shifted to a more closed conformation in the I335H variant. In agreement with an enzyme that samples more in a closed conformation, the kinetic data of the I335H variant showed a 40-fold decrease in the rate constant of substrate association (k1), a 340-fold reduction in the rate constant of substrate dissociation from the enzyme-substrate complex (k2), and a 24-fold decrease in the rate constant of product release (k5), compared to that of the wild-type. Surprisingly, the kinetic data are consistent with the mutation having a negligible effect on the reactivity of the flavin. Altogether, the data indicate that the residue at position 335 has a long-range dynamical effect on the catalytic function in PaDADH.


Subject(s)
Amino Acid Oxidoreductases , Molecular Dynamics Simulation , Amino Acid Oxidoreductases/chemistry , Catalytic Domain , Catalysis , Flavins/metabolism , Kinetics , Substrate Specificity , Binding Sites , Protein Conformation
6.
Med Res Rev ; 43(2): 293-318, 2023 03.
Article in English | MEDLINE | ID: mdl-36104980

ABSTRACT

The centrosome in animal cells is instrumental in spindle pole formation, nucleation, proper alignment of microtubules during cell division, and distribution of chromosomes in each daughter cell. Centrosome amplification involving structural and numerical abnormalities in the centrosome can cause chromosomal instability and dysregulation of the cell cycle, leading to cancer development and metastasis. However, disturbances caused by centrosome amplification can also limit cancer cell survival by activating mitotic checkpoints and promoting mitotic catastrophe. As a smart escape, cancer cells cluster their surplus of centrosomes into pseudo-bipolar spindles and progress through the cell cycle. This phenomenon, known as centrosome clustering (CC), involves many proteins and has garnered considerable attention as a specific cancer cell-targeting weapon. The kinesin-14 motor protein KIFC1 is a minus end-directed motor protein that is involved in CC. Because KIFC1 is upregulated in various cancers and modulates oncogenic signaling cascades, it has emerged as a potential chemotherapeutic target. Many molecules have been identified as KIFC1 inhibitors because of their centrosome declustering activity in cancer cells. Despite the ever-increasing literature in this field, there have been few efforts to review the progress. The current review aims to collate and present an in-depth analysis of known KIFC1 inhibitors and their biological activities. Additionally, we present computational docking data of putative KIFC1 inhibitors with their binding sites and binding affinities. This first-of-kind comparative analysis involving experimental biology, chemistry, and computational docking of different KIFC1 inhibitors may help guide decision-making in the selection and design of potent inhibitors.


Subject(s)
Benchmarking , Neoplasms , Animals , Neoplasms/pathology , Centrosome/metabolism , Binding Sites , Microtubules
7.
J Phys Chem B ; 126(51): 10844-10853, 2022 12 29.
Article in English | MEDLINE | ID: mdl-36529932

ABSTRACT

Mitochondria are the powerhouse of a cell, whose disruption due to mitochondrial pore opening can cause cell death, leading to necrosis and many other diseases. The peptidyl-prolyl cis-trans isomerase cyclophilin D (CypD) is a key player in the regulation of the mitochondrial pore. The activity of CypD can be modulated by the post-translational modification (PTM). However, the detailed mechanism of this functional modulation is not well understood. Here, we investigate the catalytic mechanism of unmodified and modified CypD by calculating the reaction free energy profiles and characterizing the function-related conformational dynamics using molecular dynamics simulations and associated analyses. Our results show that unmodified and modified CypD considerably lower the isomerization free energy barrier compared to a free peptide substrate, supporting the catalytic activity of CypD in the simulation systems. The unmodified CypD reduces the free energy difference between the cis and trans states of the peptide substrate, suggesting a stronger binding affinity of CypD toward cis, consistent with experiments. In contrast, phosphorylated CypD further stabilizes trans, leading to a lower catalytic rate in the trans-to-cis direction. The differential catalytic activities of the unmodified and phosphorylated CypD are due to a significant shift of the conformational ensemble upon phosphorylation under different functional states. Interestingly, the local flexibility is both reduced and enhanced at distinct regions by phosphorylation, which is explained by a "seesaw" model of flexibility modulation. The allosteric pathway between the phosphorylation site and a distal site displaying substantial conformational changes upon phosphorylation is also identified, which is influenced by the presence of the substrate or the substrate conformation. Similar conclusions are obtained for the acetylation of CypD using the same peptide substrate and the influence of substrate sequence is also examined. Our work may serve as the basis for the understanding of other PTMs and PTM-initiated allosteric regulations in CypD.


Subject(s)
Mitochondrial Membrane Transport Proteins , Mitochondrial Permeability Transition Pore , Peptidyl-Prolyl Isomerase F/metabolism , Mitochondrial Membrane Transport Proteins/genetics , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore/metabolism , Mitochondria , Molecular Dynamics Simulation , Protein Processing, Post-Translational
8.
J Phys Chem B ; 126(14): 2612-2620, 2022 04 14.
Article in English | MEDLINE | ID: mdl-35319195

ABSTRACT

Initiation of biological processes involving protein-ligand binding, transient protein-protein interactions, or amino acid modifications alters the conformational dynamics of proteins. Accompanying these biological processes are ensuing coupled atomic level conformational changes within the proteins. These conformational changes collectively connect multiple amino acid residues at distal allosteric, binding, and/or active sites. Local changes due to, for example, binding of a regulatory ligand at an allosteric site initiate the allosteric regulation. The allosteric signal propagates throughout the protein structure, causing changes at distal sites, activating, deactivating, or modifying the function of the protein. Hence, dynamical responses within protein structures to stimuli contain critical information on protein function. In this Perspective, we examine the description of allosteric regulation from protein dynamical responses and associated alternative and emerging computational approaches to map allosteric communication pathways between distal sites in proteins at the atomic level.


Subject(s)
Amino Acids , Proteins , Allosteric Regulation , Binding Sites , Communication , Ligands , Molecular Dynamics Simulation , Protein Conformation , Proteins/chemistry
9.
J Chem Theory Comput ; 18(2): 1173-1187, 2022 Feb 08.
Article in English | MEDLINE | ID: mdl-35048691

ABSTRACT

Allosteric regulation plays a central role in orchestrating diverse cellular processes. A prerequisite for allostery is a flexible biomolecule within which two distal sites can communicate via concerted or sequential conformational changes. We introduce a computational method to elucidate allosteric communication pathways, comprising critical allosteric residues, in biomolecules by taking advantage of conformational changes during a functional process. Conformational changes are modeled explicitly since they modulate the network of residue-residue interactions, which could propagate allosteric signals between two or more distal sites. The method implements the suboptimal path analysis in the framework of the difference contact network analysis or dCNA. The method identifies key experimentally verified allosteric residues in imidazole glycerol phosphate synthase (IGPS), a well-studied allosteric protein system. By contrast, some of the most important allosteric residues are not captured using methods that do not consider conformational changes, such as those that solely rely on examining the individual bound or unbound state of the protein. Using the dCNA path analysis along with conventional analyses, we gain several new biological insights into IGPS. Interestingly, different binding processes in the thermodynamic cycle generally use a similar group of residues in defining the allosteric communication pathways, with some residues being more specific to a certain binding process. We also observed that the fine-tuning of allosteric coupling depends on the strength of effector binding. Our results are robust against small variations of parameters and details of the network construction. The dCNA path analysis method is general and can be easily applied to diverse allosteric systems.

10.
Angew Chem Int Ed Engl ; 60(43): 23289-23298, 2021 10 18.
Article in English | MEDLINE | ID: mdl-34436811

ABSTRACT

Multi-scale calcium (Ca2+ ) dynamics, exhibiting wide-ranging temporal kinetics, constitutes a ubiquitous mode of signal transduction. We report a novel endoplasmic-reticulum (ER)-targeted Ca2+ indicator, R-CatchER, which showed superior kinetics in vitro (koff ≥2×103  s-1 , kon ≥7×106  M-1 s-1 ) and in multiple cell types. R-CatchER captured spatiotemporal ER Ca2+ dynamics in neurons and hotspots at dendritic branchpoints, enabled the first report of ER Ca2+ oscillations mediated by calcium sensing receptors (CaSRs), and revealed ER Ca2+ -based functional cooperativity of CaSR. We elucidate the mechanism of R-CatchER and propose a principle to rationally design genetically encoded Ca2+ indicators with a single Ca2+ -binding site and fast kinetics by tuning rapid fluorescent-protein dynamics and the electrostatic potential around the chromophore. The design principle is supported by the development of G-CatchER2, an upgrade of our previous (G-)CatchER with improved dynamic range. Our work may facilitate protein design, visualizing Ca2+ dynamics, and drug discovery.


Subject(s)
Calcium-Binding Proteins/metabolism , Calcium/analysis , Endoplasmic Reticulum/metabolism , Luminescent Proteins/metabolism , Animals , Calcium/metabolism , Calcium Signaling/physiology , Calcium-Binding Proteins/chemistry , HEK293 Cells , HeLa Cells , Humans , Luminescent Proteins/chemistry , Mice , Molecular Dynamics Simulation , Protein Binding , Protein Engineering , Spectrometry, Fluorescence
11.
Biochemistry ; 60(9): 711-724, 2021 03 09.
Article in English | MEDLINE | ID: mdl-33630571

ABSTRACT

Proteins are inherently dynamic, and proper enzyme function relies on conformational flexibility. In this study, we demonstrated how an active site residue changes an enzyme's reactivity by modulating fluctuations between conformational states. Replacement of tyrosine 249 (Y249) with phenylalanine in the active site of the flavin-dependent d-arginine dehydrogenase yielded an enzyme with both an active yellow FAD (Y249F-y) and an inactive chemically modified green FAD, identified as 6-OH-FAD (Y249F-g) through various spectroscopic techniques. Structural investigation of Y249F-g and Y249F-y variants by comparison to the wild-type enzyme showed no differences in the overall protein structure and fold. A closer observation of the active site of the Y249F-y enzyme revealed an alternative conformation for some active site residues and the flavin cofactor. Molecular dynamics simulations probed the alternate conformations observed in the Y249F-y enzyme structure and showed that the enzyme variant with FAD samples a metastable conformational state, not available to the wild-type enzyme. Hybrid quantum/molecular mechanical calculations identified differences in flavin electronics between the wild type and the alternate conformation of the Y249F-y enzyme. The computational studies further indicated that the alternate conformation in the Y249F-y enzyme is responsible for the higher spin density at the C6 atom of flavin, which is consistent with the formation of 6-OH-FAD in the variant enzyme. The observations in this study are consistent with an alternate conformational space that results in fine-tuning the microenvironment around a versatile cofactor playing a critical role in enzyme function.


Subject(s)
Amino Acid Oxidoreductases/chemistry , Amino Acid Oxidoreductases/metabolism , Flavins/metabolism , Phenylalanine/chemistry , Point Mutation , Pseudomonas aeruginosa/enzymology , Tyrosine/chemistry , Amino Acid Oxidoreductases/genetics , Binding Sites , Catalysis , Catalytic Domain , Kinetics , Molecular Dynamics Simulation , Mutagenesis, Site-Directed , Phenylalanine/genetics , Phenylalanine/metabolism , Protein Conformation , Tyrosine/genetics , Tyrosine/metabolism
12.
Org Biomol Chem ; 19(8): 1835-1846, 2021 03 04.
Article in English | MEDLINE | ID: mdl-33565564

ABSTRACT

Donor-π-acceptor (D-π-A) fluorophores consisting of a donor unit, a π linker, and an acceptor moiety have attracted attention in the last decade. In this study, we report the synthesis, characterization, optical properties, TD-DFT, and cytotoxicity studies of 17 near infrared (NIR) D-π-A analogs which have not been reported so far to the best of our knowledge. These fluorophores have chloroacrylic acid as the acceptor unit and various donor units such as indole, benzothiazole, benzo[e]indole, and quinoline. The fluorophores showed strong absorption in the NIR (700-970 nm) region due to their enhanced intramolecular charge transfer (ICT) between chloroacrylic acid and the donor moieties connected with the Vilsmeier-Haack linker. The emission wavelength maxima of the fluorophores were in between 798 and 870 nm. Compound 20 with a 4-quinoline donor moiety showed an emission wavelength above 1000 nm in the NIR II window. The synthesized fluorophores were characterized by 1H NMR and 13C NMR, and their optical properties were studied. Time dependent density functional theory (TD-DFT) calculations showed that the charge transfer occurs from the donor groups (indole, benzothiazole, benzo[e]indole, and quinoline) to the acceptor chloroacrylic acid moiety. Fluorophores with [HOMO] to [LUMO+1] transitions were shown to possess a charge separation character. The cytotoxicity of selected fluorophores, 4, 7, 10 and 12 was investigated against breast cancer cell lines and they showed better activity than the anti-cancer agent docetaxel.


Subject(s)
Acrylates/pharmacology , Antineoplastic Agents/pharmacology , Fluorescent Dyes/pharmacology , Heterocyclic Compounds, 2-Ring/pharmacology , Acrylates/chemical synthesis , Acrylates/radiation effects , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/radiation effects , Cell Line, Tumor , Cell Proliferation/drug effects , Density Functional Theory , Drug Screening Assays, Antitumor , Drug Stability , Fluorescent Dyes/chemical synthesis , Fluorescent Dyes/radiation effects , Heterocyclic Compounds, 2-Ring/chemical synthesis , Heterocyclic Compounds, 2-Ring/radiation effects , Humans , Light , Models, Chemical , Optical Phenomena
13.
Biochemistry ; 60(8): 597-606, 2021 03 02.
Article in English | MEDLINE | ID: mdl-33591178

ABSTRACT

The multifunctional protein p53 is the central molecular sensor of cellular stresses. The canonical function of p53 is to transcriptionally activate target genes in response to, for example, DNA damage that may trigger apoptosis. Recently, p53 was also found to play a role in the regulation of necrosis, another type of cell death featured by the mitochondrial permeability transition (mPT). In this process, p53 directly interacts with the mPT regulator cyclophilin D, the detailed mechanism of which however remains poorly understood. Here, we report a comprehensive computational investigation of the p53-cyclophilin D interaction using molecular dynamics simulations and associated analyses. We have identified the specific cyclophilin D binding site on p53 that is located at proline 151 in the DNA binding domain. As a peptidyl-prolyl isomerase, cyclophilin D binds p53 and catalyzes the cis-trans isomerization of the peptide bond preceding proline 151. We have also characterized the effect of such an isomerization and found that the p53 domain in the cis state is overall more rigid than the trans state except for the local region around proline 151. Dynamical changes upon isomerization occur in both local and distal regions, indicating an allosteric effect elicited by the isomerization. We present potential allosteric communication pathways between proline 151 and distal sites, including the DNA binding surface. Our work provides, for the first time, a model for how cyclophilin D binds p53 and regulates its activity by switching the configuration of a specific site.


Subject(s)
Cyclophilins/metabolism , DNA/metabolism , Molecular Dynamics Simulation , Proline/chemistry , Tumor Suppressor Protein p53/metabolism , Binding Sites , Catalysis , Cyclophilins/chemistry , Cyclophilins/genetics , DNA/chemistry , Humans , Proline/metabolism , Protein Domains , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/genetics
15.
Curr Opin Physiol ; 17: 269-277, 2020 Oct.
Article in English | MEDLINE | ID: mdl-33709045

ABSTRACT

Calcaium sensing receptors (CaSRs) play a central role in regulating extracellular calcium (Ca2+) homeostasis and many (patho)physiological processes. This regulation is primarily orchestrated in response to extracellular stimuli via the extracellular domain (ECD). This paper first reviews the modeled structure of the CaSR ECD and the prediction and investigation of the Ca2+ and amino acid binding sites. Several recently solved X-ray structures are then compared to support a proposed CaSR activation model involving functional cooperativity. The review also discusses recent implications for drug development. These studies provide new insights into the molecular basis of diseases and the design of therapeutic agents that target CaSR and other family C G protein-coupled receptors (cGPCRs).

16.
Acc Chem Res ; 52(12): 3455-3464, 2019 12 17.
Article in English | MEDLINE | ID: mdl-31793290

ABSTRACT

Recent advances have made all-atom molecular dynamics (MD) a powerful tool to sample the conformational energy landscape. There are still however three major challenges in the application of MD to biological systems: accuracy of force field, time scale, and the analysis of simulation trajectories. Significant progress in addressing the first two challenges has been made and extensively reviewed previously. This Account focuses on strategies of analyzing simulation data of biomolecules that also covers ways to properly design simulations and validate simulation results. In particular, we examine an approach named comparative perturbed-ensembles analysis, which we developed to efficiently detect dynamics in protein MD simulations that can be linked to biological functions. In our recent studies, we implemented this approach to understand allosteric regulations in several disease-associated human proteins. The central task of a comparative perturbed-ensembles analysis is to compare two or more conformational ensembles of a system generated by MD simulations under distinct perturbation conditions. Perturbations can be different sequence variations, ligand-binding conditions, and other physical/chemical modifications of the system. Each simulation is long enough (e.g., microsecond-long) to ensure sufficient sampling of the local substate. Then, sophisticated bioinformatic and statistical tools are applied to extract function-related information from the simulation data, including principal component analysis, residue-residue contact analysis, difference contact network analysis (dCNA) based on the graph theory, and statistical analysis of side-chain conformations. Computational findings are further validated with experimental data. By comparing distinct conformational ensembles, functional micro- to millisecond dynamics can be inferred. In contrast, such a time scale is difficult to reach in a single simulation; even when reached for a single condition of a system, it is elusive as to what dynamical motions are related to functions without, for example, comparing free and substrate-bound proteins at the minimum. We illustrate our approach with three examples. First, we discuss using the approach to identify allosteric pathways in cyclophilin A (CypA), a member of a ubiquitous class of peptidyl-prolyl cis-trans isomerase enzymes. By comparing side-chain torsion-angle distributions of CypA in wild-type and mutant forms, we identified three pathways: two are consistent with recent nuclear magnetic resonance experiments, whereas the third is a novel pathway. Second, we show how the approach enables a dynamical-evolution analysis of the human cyclophilin family. In the analysis, both conserved and divergent conformational dynamics across three cyclophilin isoforms (CypA, CypD, and CypE) were summarized. The conserved dynamics led to the discovery of allosteric networks resembling those found in CypA. A residue wise determinant underlying the unique dynamics in CypD was also detected and validated with additional mutational MD simulations. In the third example, we applied the approach to elucidate a peptide sequence-dependent allosteric mechanism in human Pin 1, a phosphorylation-dependent peptidyl-prolyl isomerase. We finally present our outlook of future directions. Especially, we envisage how the approach could help open a new avenue in drug discovery.


Subject(s)
Proteins/chemistry , Proteins/metabolism , Allosteric Regulation , Amino Acid Sequence , Animals , Humans , Molecular Dynamics Simulation , Protein Domains
17.
J Biol Chem ; 294(48): 18451-18464, 2019 11 29.
Article in English | MEDLINE | ID: mdl-31645439

ABSTRACT

Soluble guanylyl cyclase (sGC) is the main receptor for nitric oxide (NO) and a central component of the NO-cGMP pathway, critical to cardiovascular function. NO binding to the N-terminal sensor domain in sGC enhances the cyclase activity of the C-terminal catalytic domain. Our understanding of the structural elements regulating this signaling cascade is limited, hindering structure-based drug design efforts that target sGC to improve the management of cardiovascular diseases. Conformational changes are thought to propagate the NO-binding signal throughout the entire sGC heterodimer, via its coiled-coil domain, to reorient the catalytic domain into an active conformation. To identify the structural elements involved in this signal transduction cascade, here we optimized a cGMP-based luciferase assay that reports on heterologous sGC activity in Escherichia coli and identified several mutations that activate sGC. These mutations resided in the dorsal flaps, dimer interface, and GTP-binding regions of the catalytic domain. Combinations of mutations from these different elements synergized, resulting in even greater activity and indicating a complex cross-talk among these regions. Molecular dynamics simulations further revealed conformational changes underlying the functional impact of these mutations. We propose that the interfacial residues play a central role in the sGC activation mechanism by coupling the coiled-coil domain to the active site via a series of hot spots. Our results provide new mechanistic insights not only into the molecular pathway for sGC activation but also for other members of the larger nucleotidyl cyclase family.


Subject(s)
Cyclic GMP/metabolism , Molecular Dynamics Simulation , Mutation , Nitric Oxide/metabolism , Soluble Guanylyl Cyclase/genetics , Amino Acid Sequence , Animals , Catalytic Domain , Cyclic GMP/chemistry , Enzyme Activation/genetics , Humans , Kinetics , Nitric Oxide/chemistry , Protein Binding , Protein Conformation , Protein Multimerization , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism , Sequence Homology, Amino Acid , Signal Transduction , Soluble Guanylyl Cyclase/chemistry , Soluble Guanylyl Cyclase/metabolism
18.
Bioconjug Chem ; 30(10): 2647-2663, 2019 10 16.
Article in English | MEDLINE | ID: mdl-31518105

ABSTRACT

G-Quadruplex DNA has been recognized as a highly appealing target for the development of new selective chemotherapeutics, which could result in markedly reduced toxicity toward normal cells. In particular, the cyanine dyes that bind selectively to G-quadruplex structures without targeting duplex DNA have attracted attention due to their high amenability to structural modifications that allows fine-tuning of their biomolecular interactions. We have previously reported pentamethine and symmetric trimethine cyanines designed to effectively bind G-quadruplexes through end stacking interactions. Herein, we are reporting a second generation of drug candidates, the asymmetric trimethine cyanines. These have been synthesized and evaluated for their quadruplex binding properties. Incorporating a benz[c,d]indolenine heterocyclic unit increased overall quadruplex binding, and elongating the alkyl length increases the quadruplex-to-duplex binding specificity.


Subject(s)
Alkynes/chemistry , Alkynes/pharmacology , G-Quadruplexes/drug effects , Base Sequence , DNA/chemistry , DNA/genetics , Drug Design , Indoles/chemistry , Models, Molecular
19.
J Chem Inf Model ; 59(7): 3222-3228, 2019 07 22.
Article in English | MEDLINE | ID: mdl-31268315

ABSTRACT

Detailed understanding of interactions between amino acid residues is critical in using promising difference network analysis approaches to map allosteric communication pathways. Using experimental data as benchmarks, we scan values of two essential residue-residue contact parameters: the distance cutoff (dc) and the cutoff of residue separation in sequence (nc). The optimal dc = 4.5 Å is revealed, which defines the upper bound of the first shell of residue-residue packing in proteins, whereas nc is found to have little effects on performance. We also develop a new energy-based contact method for network analyses and find an equivalency between the energy network using the optimal energy cutoff ec = 1.0 kBT and the structure network using dc = 4.5 Å. The simple 4.5-Å contact method is further shown to have comparable prediction accuracy to a contact method using amino acid type-specific distance cutoffs and chemical shift prediction-based methods. This study provides necessary tools in mapping dynamics to functions.


Subject(s)
Proteins/chemistry , Amino Acid Sequence , Computational Biology/methods , Lipid Bilayers/chemistry , Models, Molecular , Molecular Dynamics Simulation , Protein Conformation , Sequence Analysis, Protein , Thermodynamics
20.
J Chem Theory Comput ; 14(11): 6026-6034, 2018 Nov 13.
Article in English | MEDLINE | ID: mdl-30351007

ABSTRACT

An automatic, multiscale, and three-dimensional (3D) summary of local configurations of the dynamics of proteins can help to discover and describe the relationships between different parts of proteins across spatial scales, including the overall conformation and 3D configurations of side chains and domains. These discoveries can improve our understanding of the function and allosteric mechanism of proteins and could potentially provide an avenue to test and improve the molecular mechanics force fields at different spatial resolutions. Many of the current methods are unable to effectively summarize shapes of 3D local configurations across all spatial scales. Here, we propose frequent substructure clustering (FSC) to fill this gap. Frequent substructure clustering of the Cß atoms of the GB3 protein identifies six clusters of co-occurring local configurations. The clusters that are localized at different regions contribute to the overall conformation, and form two anticorrelating groups. The results suggest that FSC could describe dynamical relationships between different parts of proteins by providing a 3D description of the frequently occurring local configurations at different spatial resolutions. FSC could augment the use of other methods, such as Markov state models, to study the function of subcellular processes and highlight the role of local configurations in biomolecular systems.

SELECTION OF CITATIONS
SEARCH DETAIL