Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
ACS Med Chem Lett ; 14(3): 297-304, 2023 Mar 09.
Article in English | MEDLINE | ID: mdl-36923916

ABSTRACT

Selective CDK2 inhibitors have the potential to provide effective therapeutics for CDK2-dependent cancers and for combating drug resistance due to high cyclin E1 (CCNE1) expression intrinsically or CCNE1 amplification induced by treatment of CDK4/6 inhibitors. Generative models that take advantage of deep learning are being increasingly integrated into early drug discovery for hit identification and lead optimization. Here we report the discovery of a highly potent and selective macrocyclic CDK2 inhibitor QR-6401 (23) accelerated by the application of generative models and structure-based drug design (SBDD). QR-6401 (23) demonstrated robust antitumor efficacy in an OVCAR3 ovarian cancer xenograft model via oral administration.

2.
J Comput Aided Mol Des ; 37(3): 157-166, 2023 03.
Article in English | MEDLINE | ID: mdl-36849761

ABSTRACT

The mutant KRAS was considered as an "undruggable" target for decades, especially KRASG12D. It is a great challenge to develop the inhibitors for KRASG12D which lacks the thiol group for covalently binding ligands. The discovery of MRTX1133 solved the dilemma. Interestingly, MRTX1133 can bind to both the inactive and active states of KRASG12D. The binding mechanism of MRTX1133 with KRASG12D, especially how MRTX1133 could bind the active state KRASG12D without triggering the active function of KRASG12D, has not been fully understood. Here, we used a combination of all-atom molecular dynamics simulations and Markov state model (MSM) to understand the inhibition mechanism of MRTX1133 and its analogs. The stationary probabilities derived from MSM show that MRTX1133 and its analogs can stabilize the inactive or active states of KRASG12D into different conformations. More remarkably, by scrutinizing the conformational differences, MRTX1133 and its analogs were hydrogen bonded to Gly60 to stabilize the switch II region and left switch I region in a dynamically inactive conformation, thus achieving an inhibitory effect. Our simulation and analysis provide detailed inhibition mechanism of KRASG12D induced by MRTX1133 and its analogs. This study will provide guidance for future design of novel small molecule inhibitors of KRASG12D.


Subject(s)
Molecular Dynamics Simulation , Proto-Oncogene Proteins p21(ras) , Proto-Oncogene Proteins p21(ras)/genetics , Fungal Proteins , Sulfhydryl Compounds
3.
J Comput Aided Mol Des ; 37(2): 91-105, 2023 02.
Article in English | MEDLINE | ID: mdl-36459325

ABSTRACT

G protein-coupled receptors (GPCRs) are membrane proteins constituting the largest family of drug targets. The activated GPCR binds either the heterotrimeric G proteins or arrestin through its activation cycle. Water molecules have been reported to play a role in GPCR activation. Nevertheless, reported studies are focused on the hydrophobic helical bundle region. How water molecules function in GPCR bound either G protein or arrestin is rarely studied. To address this issue, we carried out computational studies on water molecules in both GPCR/G protein complexes and GPCR/arrestin complexes. Using inhomogeneous fluid theory (IFT), we locate all possible hydration sites in GPCRs binding either to G protein or arrestin. We observe that the number of water molecules on the interaction surface between GPCRs and signal proteins are correlated with the insertion depths of the α5-helix from G-protein or "finger loop" from arrestin in GPCRs. In three out of the four simulation pairs, the interfaces of Rhodopsin, M2R and NTSR1 in the G protein-associated systems show more water-mediated hydrogen-bond networks when compared to these in arrestin-associated systems. This reflects that more functionally relevant water molecules may probably be attracted in G protein-associated structures than that in arrestin-associated structures. Moreover, we find the water-mediated interaction networks throughout the NPxxY region and the orthosteric pocket, which may be a key for GPCR activation. Reported studies show that non-biased agonist, which can trigger both GPCR-G protein and GPCR-arrestin activation signal, can result in pharmacologically toxicities. Our comprehensive studies of the hydration sites in GPCR/G protein complexes and GPCR/arrestin complexes may provide important insights in the design of G-protein biased agonists.


Subject(s)
Arrestin , Water , Arrestin/chemistry , Arrestin/metabolism , Water/metabolism , Receptors, G-Protein-Coupled/chemistry , GTP-Binding Proteins/metabolism , Rhodopsin/chemistry , Rhodopsin/metabolism
4.
Nat Commun ; 12(1): 3763, 2021 06 18.
Article in English | MEDLINE | ID: mdl-34145245

ABSTRACT

The glucagon-like peptide-1 (GLP-1) receptor is a validated drug target for metabolic disorders. Ago-allosteric modulators are capable of acting both as agonists on their own and as efficacy enhancers of orthosteric ligands. However, the molecular details of ago-allosterism remain elusive. Here, we report three cryo-electron microscopy structures of GLP-1R bound to (i) compound 2 (an ago-allosteric modulator); (ii) compound 2 and GLP-1; and (iii) compound 2 and LY3502970 (a small molecule agonist), all in complex with heterotrimeric Gs. The structures reveal that compound 2 is covalently bonded to C347 at the cytoplasmic end of TM6 and triggers its outward movement in cooperation with the ECD whose N terminus penetrates into the GLP-1 binding site. This allows compound 2 to execute positive allosteric modulation through enhancement of both agonist binding and G protein coupling. Our findings offer insights into the structural basis of ago-allosterism at GLP-1R and may aid the design of better therapeutics.


Subject(s)
Glucagon-Like Peptide 1/analogs & derivatives , Glucagon-Like Peptide 1/pharmacology , Glucagon-Like Peptide-1 Receptor/agonists , Animals , Binding Sites/physiology , CHO Cells , Cell Line , Cricetulus , Cryoelectron Microscopy , Diabetes Mellitus, Type 2/drug therapy , Enzyme Activation/drug effects , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide-1 Receptor/genetics , Glucagon-Like Peptide-1 Receptor/metabolism , Glucagon-Like Peptides/pharmacology , HEK293 Cells , Humans , Molecular Dynamics Simulation , Protein Conformation , Sf9 Cells , Spodoptera
5.
Front Chem ; 9: 666408, 2021.
Article in English | MEDLINE | ID: mdl-33937203

ABSTRACT

The over-use of antibiotics has promoted multidrug resistance and decreased the efficacy of antibiotic therapy. Thus, it is still in great need to develop efficient treatment strategies to combat the bacteria infection. The antimicrobial photodynamic therapy (aPDT) and silver nanoparticles have been emerged as effective antibacterial methods. However, the silver therapy may induce serious damages to human cells at high concentrations and, the bare silver nanoparticles may rapidly aggregate, which would reduce the antibacterial efficacy. The encapsulation of sliver by nano-carrier is a promising way to avoid its aggregation and facilitates the co-delivery of drugs for combination therapy, which does not require high concentration of sliver to exert antibacterial efficacy. This work constructed a self-assembled supermolecular nano-carrier consisting of the photosensitizers (PSs), the anti-inflammatory agent and silver. The synthesized supermolecular nano-carrier produced reactive oxygen species (ROS) under the exposure of 620-nm laser. It exhibited satisfying biocompatibility in L02 cells. And, this nano-carrier showed excellent antibacterial efficacy in Escherichia coli (E. coli) and Staphylococcus aureus (S. aureus) as indicated by bacterial growth and colony formation. Its antibacterial performance is further validated by the bacteria morphology through the scanning electron microscope (SEM), showing severely damaged structures of bacteria. To summary, the supermolecular nano-carrier TCPP-MTX-Ag-NP combining the therapeutic effects of ROS and silver may serve as a novel strategy of treatment for bacterial infection.

6.
J Colloid Interface Sci ; 593: 323-334, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33744541

ABSTRACT

In recent years, chemodynamic therapy (CDT) has gained increasing interest in cancer treatment. In contrast to photodynamic therapy and sonodynamic therapy, extrinsic excitations such as laser or ultrasound are not required in CDT. As a result, the CDT performance is not limited by the penetration depth of the external irritation. However, CDT relies heavily on hydrogen peroxide (H2O2) in the tumour microenvironment (TME). Insufficient H2O2 in the TME limits the CDT performance, and the most reported methods to produce H2O2 in the TME are dependent on oxygen supply, which is restricted by the hypoxic TME. In this study, H2O2 self-providing copper nanodots were proposed, and the drug doxorubicin (DOX) was successfully loaded to construct DOX-nanodots. Our results showed that the nanodots produced H2O2 in the weakly acidic TME due to the peroxo group and further generated the most active hydroxyl radical (OH) through the Fenton-like reaction. This process was pH-dependent and did not occur in a neutral environment. In addition to OH, the nanodots also produced singlet oxygen (1O2) and superoxide anions (O2-) in the cancer cells. The copper nanodots performed promising CDT against breast cancer in vitro and in vivo, with enhanced cell apoptosis and decreased cell proliferation. The combination of chemotherapy and CDT using DOX-nanodots further improved the therapeutic effects. The treatments showed good biocompatibility with no obvious toxicity in major tissues, possibly due to the specific OH generation in the weakly acidic TME. In summary, the H2O2 self-providing copper nanodots in combination with DOX showed promising cancer-curing effects due to the oxygen-independent and tumour-specific production of reactive oxygen species and the cooperation of chemotherapy.


Subject(s)
Breast Neoplasms , Hydrogen Peroxide , Breast Neoplasms/drug therapy , Cell Line, Tumor , Copper , Doxorubicin/pharmacology , Female , Humans , Tumor Microenvironment
7.
J Chem Theory Comput ; 17(2): 1240-1249, 2021 Feb 09.
Article in English | MEDLINE | ID: mdl-33472367

ABSTRACT

N6-Methyladenosine (m6A) is the most frequent modification in eukaryotic messenger RNA (mRNA) and its cellular processing and functions are regulated by the reader proteins YTHDCs and YTHDFs. However, the mechanism of m6A recognition by the reader proteins is still elusive. Here, we investigate this recognition process by combining atomistic simulations, site-directed mutagenesis, and biophysical experiments using YTHDC1 as a model. We find that the N6 methyl group of m6A contributes to the binding through its specific interactions with an aromatic cage (formed by Trp377 and Trp428) and also by favoring the association-prone conformation of m6A-containing RNA in solution. The m6A binding site dynamically equilibrates between multiple metastable conformations with four residues being involved in the regulation of m6A binding (Trp428, Met438, Ser378, and Thr379). Trp428 switches between two conformational states to build and dismantle the aromatic cage. Interestingly, mutating Met438 and Ser378 to alanine does not alter m6A binding to the protein but significantly redistributes the binding enthalpy and entropy terms, i.e., enthalpy-entropy compensation. Such compensation is reasoned by different entropy-enthalpy transduction associated with both conformational changes of the wild-type and mutant proteins and the redistribution of water molecules. In contrast, the point mutant Thr379Val significantly changes the thermal stability and binding capability of YTHDC1 to its natural ligand. Additionally, thermodynamic analysis and free energy calculations shed light on the role of a structural water molecule that synergistically binds to YTHDC1 with m6A and acts as the hub of a hydrogen-bond network. Taken together, the experimental data and simulation results may accelerate the discovery of chemical probes, m6A-editing tools, and drug candidates against reader proteins.


Subject(s)
Adenosine/analogs & derivatives , Nerve Tissue Proteins/chemistry , RNA Splicing Factors/chemistry , Thermodynamics , Adenosine/chemistry , Calorimetry/methods , Crystallography, X-Ray , Methylation , Molecular Conformation , Molecular Dynamics Simulation , Mutagenesis, Site-Directed , Protein Binding , Protein Domains , Water/chemistry
9.
Materials (Basel) ; 12(2)2019 Jan 16.
Article in English | MEDLINE | ID: mdl-30654431

ABSTRACT

One-dimensional, flexible yarn-shaped supercapacitors for woven cloth have the potential for use in different kinds of wearable devices. Nevertheless, the challenge that supercapacitors face is low energy density. In this paper, we present a low-cost and large-scale manufacturing method to construct a supercapacitor yarn with high power and high energy density. To construct the novel and flexible poly(3,4-ethylenedioxythiophene): poly(styrenesulfonate)⁻polyacrylonitrile (PDEOT: PSS-PAN)/Ni cotton (PNF/NiC) capacitor yarn, an electrospinning technique was initially used to wrap the polyacrylonitrile (PAN) nanofibers around the core Ni-coated yarn. The PEDOT: PSS⁻PAN nanofiber composite electrode was created using in situ deposition and H3PO4/PVA was used as a gel electrolyte. This electrode material has a yarn/nanofiber/PEDOT: PSS nanoparticle hierarchical structure, providing a high specific area and enhanced pseudocapacitance. The electrode demonstrated a high volumetric capacitance of 26.88 F·cm-3 (at 0.08 A·cm-3), an energy density of 9.56 mWh·cm-3, and a power density of 830 mW·cm-3. In addition, the PNF/NiC capacitor yarns are lightweight, highly flexible, resistant to bending fatigue, can be connected in series or parallel, and may be suitable for a variety of wearable electronic products.

10.
Elife ; 72018 10 05.
Article in English | MEDLINE | ID: mdl-30289386

ABSTRACT

Activation of heterotrimeric G proteins is a key step in many signaling cascades. However, a complete mechanism for this process, which requires allosteric communication between binding sites that are ~30 Å apart, remains elusive. We construct an atomically detailed model of G protein activation by combining three powerful computational methods: metadynamics, Markov state models (MSMs), and CARDS analysis of correlated motions. We uncover a mechanism that is consistent with a wide variety of structural and biochemical data. Surprisingly, the rate-limiting step for GDP release correlates with tilting rather than translation of the GPCR-binding helix 5. ß-Strands 1 - 3 and helix 1 emerge as hubs in the allosteric network that links conformational changes in the GPCR-binding site to disordering of the distal nucleotide-binding site and consequent GDP release. Our approach and insights provide foundations for understanding disease-implicated G protein mutants, illuminating slow events in allosteric networks, and examining unbinding processes with slow off-rates.


Subject(s)
GTP-Binding Proteins/metabolism , Guanosine Diphosphate/metabolism , Molecular Dynamics Simulation , Allosteric Regulation , Binding Sites , GTP-Binding Protein alpha Subunits, Gq-G11/chemistry , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , GTP-Binding Proteins/chemistry , Guanosine Diphosphate/chemistry , Markov Chains , Probability , Protein Structure, Secondary , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/metabolism , Thermodynamics
11.
J Chem Theory Comput ; 14(11): 5459-5475, 2018 Nov 13.
Article in English | MEDLINE | ID: mdl-30240203

ABSTRACT

Interest in atomically detailed simulations has grown significantly with recent advances in computational hardware and Markov state modeling (MSM) methods, yet outstanding questions remain that hinder their widespread adoption. Namely, how do alternative sampling strategies explore conformational space and how might this influence predictions generated from the data? Here, we seek to answer these questions for four commonly used sampling methods: (1) a single long simulation, (2) many short simulations run in parallel, (3) adaptive sampling, and (4) our recently developed goal-oriented sampling algorithm, FAST. We first develop a theoretical framework for analytically calculating the probability of discovering select states on simple landscapes, where we uncover the drastic effects of varying the number and length of simulations. We then use kinetic Monte Carlo simulations on a variety of physically inspired landscapes to characterize the probability of discovering particular states and transition pathways for each of the four methods. Consistently, we find that FAST simulations discover each target state with the highest probability, while traversing realistic pathways. Furthermore, we uncover the potential pathology that short parallel simulations sometimes predict an incorrect transition pathway by crossing large energy barriers that long simulations would typically circumnavigate. We refer to this pathology as "pathway tunneling". To protect against this phenomenon when using adaptive-sampling and FAST simulations, we introduce the FAST-string method. This method enhances sampling along the highest-flux transition paths to refine an MSMs transition probabilities and discriminate between competing pathways. Additionally, we compare the performance of a variety of MSM estimators in describing accurate thermodynamics and kinetics. For adaptive sampling, we recommend simply normalizing the transition counts out of each state after adding small pseudocounts to avoid creating sources or sinks. Lastly, we evaluate whether our insights from simple landscapes hold for all-atom molecular dynamics simulations of the folding of the λ-repressor protein. Remarkably, we find that FAST-contacts predicts the same folding pathway as a set of long simulations but with orders of magnitude less simulation time.

12.
Sci Rep ; 7(1): 16458, 2017 11 28.
Article in English | MEDLINE | ID: mdl-29184081

ABSTRACT

Neomorphic mutation R140Q in the metabolic enzyme isocitrate dehydrogenase 2 (IDH2) is found to be a driver mutation in cancers. Recent studies revealed that allosteric inhibitors could selectively inhibit IDH2/R140Q and induce differentiation of TF-1 erythroleukemia and primary human AML cells. However, the allosteric inhibition mechanism is not very clear. Here, we report the results from computational studies that AGI-6780 binds tightly with the divalent cation binding helices at the homodimer interface and prevents the transition of IDH2/R140Q homodimer to a closed conformation that is required for catalysis, resulting in the decrease of the binding free energy of NADPHs. If the allosteric inhibitor is removed, the original open catalytic center of IDH2/R140Q will gradually reorganize to a quasi-closed conformation and the enzymatic activity might recover. Unlike IDH2/R140Q, AGI-6780 locks one monomer of the wild-type IDH2 in an inactive open conformation and the other in a half-closed conformation, which can be used to explain the selectivity of AGI-6780. Our results suggest that conformational changes are the primary contributors to the inhibitory potency of the allosteric inhibitor. Our study will also facilitate the understanding of the inhibitory and selective mechanisms of AG-221 (a promising allosteric inhibitor that has been approved by FDA) for mutant IDH2.


Subject(s)
Enzyme Inhibitors/chemistry , Isocitrate Dehydrogenase/chemistry , Isocitrate Dehydrogenase/genetics , Models, Molecular , Molecular Conformation , Mutation , Alleles , Allosteric Regulation/drug effects , Allosteric Site , Amino Acid Substitution , Binding Sites , Catalytic Domain , Enzyme Inhibitors/pharmacology , Humans , Molecular Dynamics Simulation , NADP/chemistry , NADP/metabolism , Protein Binding , Protein Multimerization , Structure-Activity Relationship
13.
Chemistry ; 23(19): 4615-4624, 2017 Apr 03.
Article in English | MEDLINE | ID: mdl-28182309

ABSTRACT

Allosteric sodium in the helix bundle of a G protein-coupled receptor (GPCR) can modulate the receptor activation on the intracellular side. This phenomenon has confounded the GPCR community for decades. In this work, we present a theoretical model that reveals the mechanism of the allosteric modulation induced by sodium in the δ-opioid receptor. We found that the allosteric sodium ion exploits a distinct conformation of the key residue Trp2746.48 to propagate the modulation to helices 5 and 6, which further transmits along the helices and regulates their positions on the intracellular side. This mechanism is supported by subsequent functional assays. Remarkably, our results highlight the contrast between the allosteric effects towards two GPCR partners, the G protein and ß-arrestin, as indicated by the fact that the allosteric modulation initiated by the sodium ion significantly affects the ß-arrestin recruitment, while it alters the G protein signaling only moderately. We believe that the mechanism revealed in this work can be used to explain allosteric effects initiated by sodium in other GPCRs since the allosteric sodium is highly conserved across GPCRs.


Subject(s)
Receptors, Opioid, delta/metabolism , Sodium/metabolism , Allosteric Regulation , Allosteric Site , Humans , Molecular Dynamics Simulation , Protein Structure, Tertiary , Receptors, Opioid, delta/chemistry , Sodium/chemistry , Thermodynamics
14.
Structure ; 24(10): 1719-1728, 2016 Oct 04.
Article in English | MEDLINE | ID: mdl-27618663

ABSTRACT

The immune checkpoint receptor PD-1 and its ligand, PD-L1, have emerged as key regulators of anti-tumor immunity in humans. Recently, we reported an ultra-high-affinity PD-1 mutant, termed high-affinity consensus (HAC) PD-1, which shows superior therapeutic efficacy in mice compared with antibodies. However, the molecular details underlying the action of this agent remain incompletely understood, and a molecular view of PD-1/PD-L1 interactions in general is only beginning to emerge. Here, we report the structure of HAC PD-1 in complex with PD-L1, showing that it binds PD-L1 using a unique set of polar interactions. Biophysical studies and long-timescale molecular dynamics experiments reveal the mechanisms by which ten point mutations confer a 35,000-fold enhancement in binding affinity, and offer atomic-scale views of the role of conformational dynamics in PD-1/PD-L1 interactions. Finally, we show that the HAC PD-1 exhibits pH-dependent affinity, with pseudo-irreversible binding in a low pH setting akin to the tumor microenvironment.


Subject(s)
B7-H1 Antigen/chemistry , B7-H1 Antigen/metabolism , Point Mutation , Programmed Cell Death 1 Receptor/metabolism , Binding Sites , Crystallography, X-Ray , Humans , Hydrogen-Ion Concentration , Models, Molecular , Molecular Dynamics Simulation , Programmed Cell Death 1 Receptor/chemistry , Programmed Cell Death 1 Receptor/genetics , Protein Binding , Protein Structure, Secondary
15.
Sci Rep ; 6: 18913, 2016 Jan 08.
Article in English | MEDLINE | ID: mdl-26742690

ABSTRACT

The κ opioid receptor (κOR) is a member of G-protein-coupled receptors, and is considered as a promising drug target for treating neurological diseases. κOR selective 6'-GNTI was proved to be a G-protein biased agonist, whereas 5'-GNTI acts as an antagonist. To investigate the molecular mechanism of how these two ligands induce different behaviors of the receptor, we built two systems containing the 5'-GNTI-κOR complex and the 6'-GNTI-κOR complex, respectively, and performed molecular dynamics simulations of the two systems. We observe that transmembrane (TM) helix 6 of the κOR rotates about 4.6(o) on average in the κOR-6'-GNTI complex. Detailed analyses of the simulation results indicate that E297(6.58) and I294(6.55) play crucial roles in the rotation of TM6. In the simulation of the κOR-5'-GNTI system, it is revealed that 5'-GNTI can stabilize TM6 in the inactive state form. In addition, the kink of TM7 is stabilized by a hydrogen bond between S324(7.47) and the residue V69(1.42) on TM1.


Subject(s)
Analgesics, Opioid/chemistry , Guanidines/chemistry , Morphinans/chemistry , Naltrexone/analogs & derivatives , Narcotic Antagonists/chemistry , Receptors, Opioid, kappa/chemistry , Sodium/chemistry , Allosteric Regulation , Amino Acid Motifs , Cations, Monovalent , Gene Expression , Humans , Hydrogen Bonding , Ligands , Molecular Docking Simulation , Molecular Dynamics Simulation , Mutation , Naltrexone/chemistry , Protein Interaction Domains and Motifs , Protein Structure, Secondary , Receptors, Opioid, kappa/agonists , Receptors, Opioid, kappa/antagonists & inhibitors , Receptors, Opioid, kappa/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics
16.
Bioorg Med Chem ; 23(24): 7735-42, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26643219

ABSTRACT

A series of novel arylpiperazine derivatives as α1A/1D-adrenergic receptors (AR) subtype selective antagonists were designed, synthesized and evaluated for their antagonistic activities towards α1-ARs (α1A, α1B, and α1D). Compounds 9, 12, 13, 15, 17, 18, 21, 22, 25 and 26 exerted strong antagonistic effects on α1A and/or α1D subtypes over α1B in vitro. SAR analysis indicated that chloride at the ortho-phenyl position for compound 17 was beneficial for the highest α1A/D-AR sub-selectivity. Moreover, molecular docking study of compound 17 with the homology-modeled α1-ARs (α1A, α1B, and α1D) structures exhibited differences of key amino resides in the docking pocket which may influence the subtype selectivity. ILE 193 of α1A was validated as the key residues for binding ligand. This work provides useful information for finding more new potential drugs in clinic in treating benign prostatic hyperplasia (BPH).


Subject(s)
Adrenergic alpha-1 Receptor Antagonists/chemistry , Adrenergic alpha-1 Receptor Antagonists/pharmacology , Piperazines/chemistry , Piperazines/pharmacology , Receptors, Adrenergic, alpha-1/metabolism , Humans , Male , Molecular Docking Simulation , Prostatic Hyperplasia/drug therapy , Prostatic Hyperplasia/metabolism , Structure-Activity Relationship
17.
Phys Chem Chem Phys ; 17(26): 16886-93, 2015 Jul 14.
Article in English | MEDLINE | ID: mdl-26060853

ABSTRACT

Amyloid beta (Aß) peptides are small cleavage products of the amyloid precursor protein. Aggregates of Aß peptides are thought to be linked with Alzheimer's and other neurodegenerative diseases. Strategies aimed at inhibiting amyloid formation and promoting Aß clearance have been proposed and investigated in in vitro experiments and in vivo therapies. A recent study indicated that a novel affibody protein ZAß3, which binds to an Aß40 monomer with a binding affinity of 17 nM, is able to prevent the aggregation of Aß40. However, little is known about the energetic contribution of each residue in ZAß3 to the formation of the (ZAß3)2:Aß complex. To address this issue, we carried out unbiased molecular dynamics simulations and molecular mechanics Poisson-Boltzmann surface area calculations. Through the per-residue decomposition scheme, we identified that the van der Waals interactions between the hydrophobic residues of (ZAß3)2 and those at the exterior and interior faces of Aß are the main contributors to the binding of (ZAß3)2 to Aß. Computational alanine scanning identified 5 hot spots, all residing in the binding interface and contributing to the binding of (ZAß3)2 to Aß through the hydrophobic effect. In addition, the amide hydrogen bonds in the 4-strand ß-sheet and the π-π stacking were also analyzed. Overall, our study provides a theoretical basis for future experimental improvement of the ZAß3 peptide binding to Aß.


Subject(s)
Amyloid beta-Peptides/chemistry , Recombinant Fusion Proteins/chemistry , Hydrogen Bonding , Molecular Dynamics Simulation , Protein Binding , Thermodynamics
18.
Sci Rep ; 5: 8066, 2015 Jan 28.
Article in English | MEDLINE | ID: mdl-25628267

ABSTRACT

The corticotropin releasing factors receptor-1 and receptor-2 (CRF1R and CRF2R) are therapeutic targets for treating neurological diseases. Antagonists targeting CRF1R have been developed for the potential treatment of anxiety disorders and alcohol addiction. It has been found that antagonists targeting CRF1R always show high selectivity, although CRF1R and CRF2R share a very high rate of sequence identity. This has inspired us to study the origin of the selectivity of the antagonists. We have therefore built a homology model for CRF2R and carried out unbiased molecular dynamics and well-tempered metadynamics simulations for systems with the antagonist CP-376395 in CRF1R or CRF2R to address this issue. We found that the side chain of Tyr(6.63) forms a hydrogen bond with the residue remote from the binding pocket, which allows Tyr(6.63) to adopt different conformations in the two receptors and results in the presence or absence of a bottleneck controlling the antagonist binding to or dissociation from the receptors. The rotameric switch of the side chain of Tyr356(6.63) allows the breaking down of the bottleneck and is a perquisite for the dissociation of CP-376395 from CRF1R.


Subject(s)
Receptors, Corticotropin-Releasing Hormone/metabolism , Aminopyridines/chemistry , Aminopyridines/metabolism , Binding Sites , Humans , Hydrogen Bonding , Molecular Dynamics Simulation , Protein Structure, Tertiary , Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors , Tyrosine/chemistry
19.
J Phys Chem B ; 118(51): 14737-44, 2014 Dec 26.
Article in English | MEDLINE | ID: mdl-25453446

ABSTRACT

Experiments have revealed that in the ß(2) adrenergic receptor (ß(2)AR)-Gs protein complex the α subunit (Gαs) of the Gs protein can adopt either an "open" conformation or a "closed" conformation. In the "open" conformation the Gs protein prefers to bind to the ß(2)AR, while in the "closed" conformation an uncoupling of the Gs protein from the ß(2)AR occurs. However, the mechanism that leads to such different behaviors of the Gs protein remains unclear. Here, we report results from microsecond molecular dynamics simulations and community network analysis of the ß(2)AR-Gs complex with Gαs in the "open" and "closed" conformations. We observed that the complex is stabilized differently in the "open" and "closed" conformations. The community network analysis reveals that in the "closed" conformation there exists strong allosteric communication between the ß(2)AR and Gßγ, mediated by Gαs. We suggest that such high information flows are necessary for the Gs protein uncoupling from the ß(2)AR.


Subject(s)
GTP-Binding Protein alpha Subunits, Gs/chemistry , Receptors, Adrenergic, beta-2/chemistry , Allosteric Regulation , Molecular Dynamics Simulation
20.
J Phys Chem B ; 118(37): 10863-73, 2014 Sep 18.
Article in English | MEDLINE | ID: mdl-25166739

ABSTRACT

G-protein-coupled receptors (GPCRs) are integral membrane proteins that mediate cellular response to an extensive variety of extracellular stimuli. Studies of rhodopsin, a prototype GPCR, have suggested that water plays an important role in mediating the activation of family A GPCRs. However, our understanding of the function of water molecules in the GPCR activation is still rather limited because resolving the functional water molecules solely based on the results from existing experiments is challenging. Using all-atom molecular dynamics simulations in combination with inhomogeneous fluid theory, we identify in this work the positioning of functional water molecules in the inactive state, the Meta II state, and the constitutive active state of rhodopsin, basing on the thermodynamic signatures of the water molecules. We find that one hydration site likely functions as a switch to regulate the distance between Glu181 and the Schiff base in the rhodopsin activation. We observe that water molecules adjacent to the "NpxxY" motif are not as stable in the Meta II state as in the inactive state as indicated by the thermodynamics signatures, and we rationalize that the behaviors of these water molecules are closely correlated with the rearrangement of the water-mediated hydrogen-bond network in the "NPxxY" motif, which is essential for mediating the activation of the receptor. We thereby propose a hypothesis of the water-mediated rhodopsin activation pathway.


Subject(s)
Rhodopsin/chemistry , Water/chemistry , Amino Acid Motifs , Hydrogen Bonding , Molecular Dynamics Simulation , Protein Structure, Tertiary , Rhodopsin/metabolism , Schiff Bases/chemistry , Thermodynamics
SELECTION OF CITATIONS
SEARCH DETAIL
...